EP3452493A1 - Molécules d'acide nucléique et leurs utilisations - Google Patents

Molécules d'acide nucléique et leurs utilisations

Info

Publication number
EP3452493A1
EP3452493A1 EP17725524.7A EP17725524A EP3452493A1 EP 3452493 A1 EP3452493 A1 EP 3452493A1 EP 17725524 A EP17725524 A EP 17725524A EP 3452493 A1 EP3452493 A1 EP 3452493A1
Authority
EP
European Patent Office
Prior art keywords
nucleic acid
norovirus
sequence
artificial nucleic
gii
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP17725524.7A
Other languages
German (de)
English (en)
Inventor
Susanne RAUCH
Kim Ellen SCHMIDT
Benjamin Petsch
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Curevac SE
Original Assignee
Curevac AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Curevac AG filed Critical Curevac AG
Publication of EP3452493A1 publication Critical patent/EP3452493A1/fr
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/125Picornaviridae, e.g. calicivirus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5258Virus-like particles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/16011Caliciviridae
    • C12N2770/16034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the present invention is directed to an artificial nucleic acid and to polypeptides suitable for use in treatment or prophylaxis of ID an infection with Norovirus or a disorder related to such an infection.
  • the present invention concerns a Norovirus vaccine.
  • the present invention is directed to an artificial nucleic acid, polypeptides, compositions and vaccines comprising the artificial nucleic acid or the polypeptides.
  • the invention further concerns a method of treating or preventing a disorder or a disease, first and second medical uses of the artificial nucleic acid, polypeptides, compositions and vaccines.
  • the invention is directed to a kit, particularly to a kit of parts, comprising the artificial nucleic acid, polypeptides, compositions and 15 vaccines.
  • Noroviruses also known as Norwalk-like viruses or Norwalk viruses
  • Norwalk-like viruses or Norwalk viruses are positive sense, single-stranded RNA Calciviruses (Sarvestani, Soroush I, et al. "Norovirus Infection: Replication, Manipulation of Host, and Interaction with the Host Immune Response.” Journal of Interferon FJ Cytokine Research 36.4 (20 ⁇ 6):2 ⁇ 5-225), containing a non-segmented RNA genome.
  • the virusD genome is organized in three open reading frames, of which the 5' proximal DRF encodes a large polyprotein that is cleaved into non-structural proteins; the minor capsid protein VP2 is encoded by 0RF3 and the major capsid protein VPI is encoded by DRF2 (Karst et al.. Cell Host Microbe II; I5(B):BG8-8D, 2014; Robilotti E, Deresinski S, Pinsky BA. 2015. Norovirus. Clin Microbiol Rev 28:134-164).
  • Noroviruses are classified into five genogroups (Gl - GV), and are further subdivided into genotypes based on the capsid sequence (Zheng, Du-Ping, et al. "Norovirus classification and proposed strain nomenclature.” Virology 34G.2 (2006):312-323; Kroneman. A., et al. "An automated genotyping tool for enteroviruses and noroviruses.” Journal of Clinical Virology 51.2 (20II):I2I-I25). Usually viruses of genogroups I, II are known to infect humans (Ramani, Sasirekha, Robert L. Atmar, and Mary K. Estes.
  • Infections with Noroviruses are generally self-limiting in healthy adults, displaying typical symptoms including non-bloody5 diarrhea, vomiting, nausea and abdominal cramps.
  • infections can be severe and even fatal (Glass, Roger I., Omesh D. Parashar, and Mary K. Estes. "Norovirus gastrDEnteritis.” NEW England Journal of MBdicine 361.18 (20D9):I77G-I785).
  • Nongastrointestinal- related illness, including neuradevelopmental disorders have also been reported after Norovirus infection. (Sarvestani, Soroush I, et al. "Norovirus Infection: Replication, Manipulation of Host, and Interaction with the Host Immune Response.” Journal of Interferon 5 Cytokine Research 36.4 (20l6):2l5-225).
  • NoV has been shown to be the cause of the majority of nonbacterial gastroenteritis epidemics, resulting in a huge economic burden. In the US, the cost of NoV-associated
  • Norovirus infections there is no specific treatment of Norovirus infections. Therapy is limited to curing the symptoms caused by the infection.
  • the underlying objEct of the present invention is therefore to provide a Norovirus vaccine. It is a further preferred object of the invention to provide a Norovirus vaccine, which may be produced at an industrial scale. A further object of the present invention is the provision of a storage-stable Norovirus vaccine. Further object of the underlying invention is to provide mRNA sequences5 coding for antigenic peptides or proteins derived from a protein of a Norovirus or a fragment or variant thereof for the use as a vaccine for prophylaxis or treatment of Norovirus infections. Furthermore, it is the object of the present invention to provide an effective Norovirus vaccine which can be stored without cold chain and which enables rapid and scalable vaccine production.
  • the object underlying the present invention is solved by the claimed subject-matter.
  • the objects underlying the0 present invention are solved according to a first aspect by an inventive by providing an artificial nucleic acid comprising at least one coding region encoding at least one polypeptide derived from a Norovirus, aod/or a fragment or variant thereof. Definitions:
  • the adaptive immune response is typically understood to be an antigen-specific response of the immune system. Antigen specificity allows for the generation of responses that are tailored to specific pathogens or pathogen- infected cells. The ability to mount these tailored responses is usually maintained in the body by "memory cells". Should a pathogen infect the body more than once, these specific memory cells are used to quickly eliminate it.
  • the first step of an adaptive immune response is the activation of naive antigen-specific T cells or different immune cells able to induce an antigen-specific immune response by antigen-presenting cells. This occurs in the lymphoid tissues and organs through which naive T cells are constantly passing.
  • dendritic cells The three cell types that may serve as antigen-presenting cells are dendritic cells, macrophages, and B cells. Each of these cells has a distinct function in eliciting immune responses.
  • Dendritic cells may take up antigens by phagocytosis and macropinocytosis and may become stimulated by contact with e.g. a foreign antigen to migrate to the local lymphoid tissue, where they differentiate into mature dendritic cells.
  • Macrophages ingest particulate antigens such as bacteria and are induced by infectious agents or other appropriate stimuli to express MHC molecules.
  • the unique ability of B cells to bind and internalize soluble protein antigens via their receptors may also be important to induce T cells.
  • MHC-molecules are, typically, responsible for presentation of an antigen to T-cells. Therein, presenting the antigen on MHC molecules leads to activation of T cells which induces their proliferation and differentiation into armed effector T cells.
  • effector T cells The most important function of effector T cells is the killing of infected cells by CD8+ cytotoxic T cells and the activation of macrophages by Thl cells which together make up cell-mediated immunity, and the activation of B cells by both ThZ and Thl cells to produce different classes of antibody, thus driving the humoral immune response.
  • T cells recognize an antigen by their T cell receptors which do not recognize and bind the antigen directly, but instead recognize short peptide fragments e.g. of pathogen-derived protein antigens, e.g. so- called epitopes, which are bound to MHC molecules on the surfaces of other cells.
  • the adaptive immune system is essentially dedicated to eliminate or prevent pathogenic growth. It typically regulates the adaptive immune response by providing the vertebrate immune system with the ability to recognize and remember specific pathogens (to generate immunity), and to mount stronger attacks each time the pathogen is encountered.
  • the system is highly adaptable because of somatic hypermutation (a process of accelerated somatic mutations), and V(D)J recombination (an irreversible genetic recombination of antigen receptor gene segments). This mechanism allows a small number of genes to generate a vast number of different antigen receptors, which are then uniquely expressed on each individual lymphocyte.
  • Adjuvant/ adjuvant component An adjuvant OP an adjuvant component in the broadest sense is typically a pharmacological and/ or immunological agent that may modify, e.g. enhance, the effect of other agents, such as a drug or vaccine. It is to be interpreted in a broad sense and refers to a broad spectrum of substances. Typically, these substances are able to increase the immunogenic ⁇ of antigens.
  • adjuvants may be recognized by the innate immune systems and, e.g., may elicit an 5 innate immune response. "Adjuvants” typically do not elicit an adaptive immune response. Insofar, “adjuvants” do not qualify as antigens. Their mode of action is distinct from the effects triggered by antigens resulting in an adaptive immune response.
  • Antigen refers typically to a substance which may be recognized by the immune system, preferably by the adaptive immune system, and is capable of triggering an antigen-specific immune response, e.g. by ID formation of antibodies and/ or antigen-specific T cells as part of an adaptive immune response.
  • an antigen may be or may comprise a peptide or protein which may be presented by the MHC to T-cells.
  • an antigen may be the product of translation of a provided nucleic acid molecule, preferably an m NA as defined herein.
  • fragments, variants and derivatives of peptides and proteins comprising at least one epitope are understood as antigens.
  • tumour antigens and pathogenic antigens as defined herein are particularly preferred.
  • Artificial nucleic acid molecule An "artificial nucleic acid molecule” or “artificial nucleic acid” may typically be understood to be a nucleic acid molecule, e.g. a DNA or an RNA that does not occur naturally. In other words, an artificial nucleic acid molecule may be understood as a non-natural nucleic acid molecule. Such nucleic acid molecule may be non-natural due to its individual sequence (which does not occur naturally) and/or due to other modifications, e.g. structural modifications of nucleotides whichD do not occur naturally.
  • An artificial nucleic acid molecule may be a DNA molecule, an RNA molecule or a hybrid-molecule comprising DNA and RNA portions.
  • artificial nucleic acid molecules may be designed and/or generated by genetic engineering methods to correspond to a desired artificial sequence of nucleotides (heterologous sequence).
  • an artificial sequence is usually a sequence that may not occur naturally, i.e. it differs from the wild type sequence by at least one nucleotide.
  • wild type may be understood as a sequence occurring in nature.
  • artificial nucleic acid5 molecule is not restricted to mean “one single molecule” but is, typically, understood to comprise an ensemble of identical molecules. Accordingly, it may relate to a plurality of identical molecules contained in an aliquot.
  • a bicistronic or multicistronic nucleic acid or RNA is typically a nucleic acid or an RNA, preferably an mRNA, that typically may have two (bicistronic) or more (multicistronic) codingD regions.
  • a coding region in this context is a sequence of codons that is translatable into a peptide or protein.
  • Carrier/polymeric carrier A carrier in the context of the invention may typically be a compound that facilitates transport and/ or complexation of another compound (cargo).
  • a polymeric carrier is typically a carrier that is formed of a polymer.
  • a carrier may be associated to its cargo by covalent or non-covalent interaction.
  • a carrier may transport nucleic acids, e.g. RNA or DNA, to the5 target cells.
  • the carrier may - for some embodiments - be a cationic component.
  • Complexation and Formulation According to a preferred embodiment, the at least one m NA of the inventive composition may be complexed with lipids to form one or more liposomes, lipoplexes, or lipid nanoparticles. Therefore, in one embodiment, the inventive composition comprises liposomes, lipoplexes, and/or lipid nanoparticles comprising the at least one mRNA.
  • Lipid-based formulations have been increasingly recognized as one of the mast promising delivery systems for RNA due to their biocompatibility and their ease of large-scale production.
  • Cationic lipids have been widely studied as synthetic materials for delivery of RNA.
  • nucleic acids are condensed by cationic lipids to form lipid/ nucleic acid complexes known as lipoplexes.
  • lipoplexes are able to protect genetic material from the action of nucleases and deliver it into cells by interacting with the negatively charged cell membrane.
  • Lipoplexes can be prepared by directly mixing positively charged lipids
  • liposomes consist of a lipid bilayer that can be composed of cationic, anionic, or neutral (phospho)lipids and cholesterol, which encloses an aqueous core. Both the lipid bilayer and the aqueous space can incorporate hydrophobic or hydrophilic compounds, respectively. Liposome characteristics and behaviour in vivo can be modified by addition of a hydrophilic polymer coating, e.g. polyethylene glycol (PEG), to the liposome surface to confer steric stabilization. Furthermore, liposomes can be used for specific targeting by attaching ligands (e.g., antibodies, peptides,
  • ligands e.g., antibodies, peptides,
  • Liposomes are colloidal lipid-based and surfactant-based delivery systems composed of a phospholipid bilayer surrounding an aqueous compartment. They may present as spherical vesicles and can range in size from 20 nm to a few microns. Cationic lipid-based liposomes are able to complex with negatively charged nucleic acids via electrostatic interactions, resulting in complexes that offer biocompatibility, low toxicity, and the possibility of the large-scale production required for in vivo clinicalD applications.
  • Liposomes can fose with the plasma membrane for uptake; once inside the cell, the liposomes are processed via the endocytic pathway and the genetic material is then released from the endnsome/carrier into the cytoplasm. Liposomes have long been perceived as drug delivery vehicles because of their superior biocompatibility, given that liposomes are basically analogs of biological membranes, and can be prepared from both natural and synthetic phospholipids (Int J Nanomedicine.2014; 3:1833-1843).
  • Cationic liposomes have been traditionally the most commonly used non-viral delivery systems for oligonucleotides, including plasmid DNA, antisense oligos, and siRNA/small hairpin RNA-shRNA).
  • Cationic lipids such as DDTAP, (l,2-dioleoyl-3- trimethylammonium-propane) and DOTMA (N-[l-(2,3-dmleoyloxy)propyl]-N,N,N-trimethyl-ammonium methyl sulfate) can form complexes or lipoplexes with negatively charged nucleic acids to form nanoparticles by electrostatic interactioo, providing highD in vitro transfection efficiency .
  • DDTAP l,2-dioleoyl-3- trimethylammonium-propane
  • DOTMA N-[l-(2,3-dmleoyloxy)propyl]-N,N,N-trimethyl-ammonium methyl
  • neutral lipid-based nanoliposomes for RNA delivery as e.g. neutral 1,2-diolBoyl-sn- glycero-3-phosphatidylchaline (DOPC)-based nanoliposomes were developed. (Adv Drug Deliv Rev.2DI4 Feb; 66:110-116).
  • DOPC 1,2-diolBoyl-sn- glycero-3-phosphatidylchaline
  • the at least one mRNA of the inventive compositioo is complexed with cationic lipids and/ or neutral lipids and thereby forms liposomes, lipid nanoparticles, lipoplexes or neutral lipid-based nanoliposomes.
  • Cationic campanent or catipnic compound typically refers to a charged molecule, which is positively charged (cation) at a pH value typically from I to 3, preferably at a pH value of or below 9 (e.g. from 5 to 9), of or below 8 (e.g. from 5 to 8), of or below 7 (e.g. from 5 to 7), most preferably at a physiological pH, e.g. from 7.3 to 7.4.
  • a cationic component may be any positively charged compound or polymer, preferably a cationic peptide or
  • a cationic peptide, protein, polysaccharide, lipid or polymer according to the present invention is positively charged under physiological conditions, particularly under physiological salt conditions of the cell in vivo.
  • a "cationic peptide or protein” may contain at least one positively charged amino acid, or more than one positively charged amino acid, e.g. selected from Arg, His, Lys or Drn. Accordingly, "polycationic" components or compounds are also within the scope exhibiting more than one positive
  • a 5'-cap is an entity, typically a modified nucleotide entity, which generally "caps" the S'-end of a mature mRNA.
  • a 5'-cap may typically be formed by a modified nucleotide, particularly by a derivative of a guanine nucleotide.
  • the 5'-cap is linked to the 5'-terminus via a 5'-5'-triphosphate linkage.
  • a 5'-cap may be methylated, e.g. m7GpppN, wherein N is the terminal
  • 5'cap structures include glyceryl, inverted deoxy abasic residue (moiety), 4',5' methylene nucleotide, l-(beta-D-erythrofuranosyl) nucleotide, 4'-thio nucleotide, carbocyclic nucleotide, 1,5-anhydrohexitol nucleotide, L-nucleotides, alpha-nucleotide, modified base nucleotide, threo-pentofuranosyl nucleotide, acyclic 3',4'-seco nucleotide, acyclic 3,4-dihydroxybutyl nucleotide, acyclic 3,5 dihydroxypentyl nucleotide.3'-3'-inverted nucleotide moiety, 3'-3'-inverted nucleotide moiety, 3'-3'-inverted nucleotide moiety, 3'-3'-inverted nucleo
  • a cap analogue refers to a non-polymerizable di-nucleotide that has cap functionality in that it facilitates translation or localization, and/or prevents degradation of a nucleic acid molecule, particularly of an RNA molecule, when5 incorporated at the 5' end of the nucleic acid molecule.
  • Non-polymerizable means that the cap analogue will be incorporated only at the 5'-terminus because it does not have a 5' triphosphate and therefore cannot be extended in the 3' direction by a template- dependent polymerase, particularly, by template-dependent RNA polymerase.
  • Cap analogues include, but are not limited to, a chemical structure selected from the group consisting of m7GpppG, m7GpppA,D m7GpppC; unmethylated cap analogues (e.g., GpppG); dimethylated cap analogue (e.g., m2.7GpppG), trimethylated cap analogue (e.g., m2,2,7GpppG), dimethylated symmetrical cap analogues (e.g., m7Gpppm7G), or anti reverse cap analogues (e.g., ARCA; m7,2'0meGpppG.
  • unmethylated cap analogues e.g., GpppG
  • dimethylated cap analogue e.g., m2.7GpppG
  • trimethylated cap analogue e.g., m2,2,7GpppG
  • dimethylated symmetrical cap analogues e.g.,
  • a 5'-cap is typically a a modified nucleotide (cap analogue), particularly a guanine nucleotide, added to the 5' end of a nucleic acid molecule, particularly of an RNA molecule, e.g. an mRNA molecule.
  • the 5'- cap is added using a 5'-5'-triphosphate linkage (also named m7GpppN).
  • 5'- cap structures include glyceryl, inverted deoxy abasic residue (moiety), 4',5' methylene nucleotide, l-(beta-D-erythrofuranosyl) nucleotide, 4'-thio nucleotide, carbocyclic nucleotide.
  • 1,5-anhydrohexitol nucleotide L-nucleotides, alpha-nucleatide, modified base nucleotide, thren-pentofuranosyl nucleotide, acyclic 3',4'-seco nucleotide, acyclic 3,4-dihydroxybutyl nucleotide, acyclic 3,5 dihydroxypentyl nucleotide, 3'-3'-inverted nucleotide moiety, 3'-3'-inverted abasic moiety, 3'-2'-inverted nucleotide moiety, 3'-2'-inverted abasic moiety, 1,4-butanediol phosphate, 3'- phasphoramidate, hexylphosphate, aminohexyl phosphate, 3'-phasphate, 3'phosphorothioate, phosphorodithioate, or bridging or non-bridging methylphosphonate moiety.
  • modified 5'-cap structures may be used in the context of the present invention to modify the mRNA sequence of the inventive composition.
  • Further modified 5'-cap structures which may be used in the context of the present invention are cap I (additional methylation of the ribose of the adjacent nucleotide of m7GpppN), cap2 (additional methylation of the ribose of the 2nd nucleotide downstream of the m7GpppN), cap3 (additional methylation of the ribose of the 3rd nucleotide downstream of the m7GpppN), cap4 (additional methylation of the ribose of the 4th nucleotide downstream of the m7GpppN), ARCA (anti-reverse cap analogue), modified ARCA (e.g.
  • phosphothioate modified ARCA inosine, Nl-methyl-guanosine, 2'-fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido-guanosine.
  • a 5' cap structure may also be formed in chemical RNA synthesis or RNA in vitro transcription (co-transcriptional capping) using cap analogues, or a cap structure may be formed in vitro using capping enzymes (e.g., commercially available capping kits).
  • a cap structure e.g., capO or capl
  • Ghemical synthesis of nucleic acids Chemical synthesis of relatively short fragments of oligonucleotides with defined chemical structure provides a rapid and inexpensive access to custom-made oligonucleotides of any desired sequence. Whereas enzymes synthesize DNA and RNA only in the 5' to 3' direction, chemical oligonucleotide synthesis does not have this limitation, although it is most often carried out in the opposite, i.e. the 3' to 5' direction. Currently, the process is implemented as solid-phase synthesis using the phosphoramidite methnd and phosphoramidite building blocks derived from protected nucleosides (A, C, G, and U), or chemically modified nucleosides.
  • the building blocks are sequentially coupled to the growing oligonucleotide chain on a solid phase in the order required by the sequence of the product in a fully automated process.
  • the product is released from the solid phase to the solution, deprotected, and collected.
  • the occurrence of side reactions sets practical limits for the length of synthetic oligonucleotides (up to about 20D nucleotide residues), because the number of errnrs increases with the length of the oligonucleotide being synthesized.
  • Products are often isolated by HPLC to obtain the desired oligonucleotides in high purity.
  • oligonucleotides find a variety of applications in molecular biology and medicine. They are most commonly 5 used as antisense oligonucleotides, small interfering RNA, primers for DNA sequencing and amplification, probes for detecting complementary DNA or RNA via molecular hybridization, tools for the targeted introduction of mutations and restriction sites, and for the synthesis of artificial genes. Moreover, long-chain DNA molecules and long-chain RNA molecules may be chemically synthetized and used in the context of the present invention.
  • Cellular immunity relates typically to the activation of macrophages, natural killer cells (NK). antigen-specific cytotoxic T-lymphncytes, and the release of various cytokines in response to an antigen.
  • cellular immunity is not based on antibodies, but on the activation of cells of the immune system.
  • a cellular immune response may be characterized e.g. by activating antigen-specific cytotoxic T-lymphocytes that are able to induce apoptosis in cells, e.g. specific immune cells like dendritic cells or other cells, displaying epitopes of foreign antigens on
  • Such cells may be virus-infected or infected with intracellular bacteria, or cancer cells displaying tumor antigens.
  • a cloning site is typically understood to be a segment of a nucleic acid molecule, which is suitable for insertion of a nucleic acid sequence, e.g., a nucleic acid sequence comprising a coding region. Insertion may be performed by any molecular biological method known to the one skilled in the art, e.g. by restriction and ligation.
  • a cloning site typically comprises Dne or more restriction enzyme recognition sites (restriction sites).
  • Dne or more restrictions sites may be recognized by restriction enzymes which cleave the DNA at these sites.
  • a cloning site which comprises more than one restriction site may also5 be termed a multiple cloning site (MCS) or a polylinker.
  • Coding region, coding sequence is typically a sequence of several nucleotide triplets, which may be translated into a peptide or protein.
  • a coding region preferably contains a start codon, i.e. a combination of three subsequent nucleotides coding usually for the amino acid methionine (ATG), at its 5'-end and a subsequent region whichD usually exhibits a length which is a multiple of 3 nucleotides.
  • a coding region is preferably terminated by a stop-codon (e.g., TAA, TAG, TGA). Typically, this is the only stop-codon of the coding region.
  • a coding region in the context of the present invention is preferably a nucleotide sequence, consisting of a number of nucleotides that may be divided by three, which starts with a start codon (e.g. ATG) and which preferably terminates with a stop codon (e.g., TAA, TGA, or TAG).
  • the coding region may be isolated or it may be incorporated in a longer nucleic acid sequence, for example in a vector or an mRNA.
  • a coding region may also be termed "protein coding region", "coding sequence", “CDS”, "open reading frame” or "DRF".
  • nucleic acid which is derived from (another) nucleic acid, shares at least 50%, preferably at least 60%, preferably at least 70%, more preferably at least 75%, more preferably at least 80%, more preferably at least 85%, even more preferably at least 90%, even more preferably at least 95%, and particularly preferably at least 98% sequence identity with the nucleic acid from which it is derived.
  • "derived from” means having in increasing order of preference at least 50%, 51%, 52%, 53%, 54%, 55%, 56%. 57%, 58%, 58%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%. 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more sequence identity to the sequences as represented by SEQ 10 NOs: 1-39690. 39713-39746.
  • sequence identity is typically calculated for the same types of nucleic acids, i.e. for DNA sequences or for RNA sequences.
  • a DNA is “derived from” an RNA or if an RNA is “derived from” a DNA.
  • the RNA sequence is converted into the corresponding DNA sequence (in particular by replacing the uracils (U) by thymidines (T) throughout the sequence) or, vice versa, the DNA sequence is converted into the corresponding RNA sequence (in particular by replacing the thymidines (T) by uracils (U) throughout the sequence).
  • sequence identity of the DNA sequences or the sequence identity of the RNA sequences is determined.
  • a nucleic acid "derived from” a nucleic acid also refers to nucleic acid, which is modified in comparison to the nucleic acid from which it is derived, e.g. in order to increase RNA stability even further and/ or to prolong and/or increase protein production. It goes without saying that such modifications are preferred, which do not impair RNA stability, e.g. in comparison to the nucleic acid from which it is derived.
  • Different Noro virus The term "different Noro virus” in the context of the invention has to be understood as the difference between at least two respective Noroviruses, wherein the difference is manifested on the RNA genome of the respective different virus.
  • different Norovirus has to be understood as genetically “different Norovirus”.
  • said (genetically) different Noroviruses express at least one different protein or peptide, wherein the at least one different protein or peptide preferably differs in at least one amino acid.
  • DNA is the usual abbreviation for deoxyribonucleic acid. It is a nucleic acid molecule, i.e. a polymer consisting of nucleotides.
  • nucleotides are usually deoxy-adenosine-monophosphate, deoxy-thymidine-monophosphate, deoxy-guanasine- monaphasphate and deoxy-cytidine-monophosphate monomers which are - by themselves - composed of a sugar moiety (deoxyribose), a base moiety and a phosphate moiety, and polymerize by a characteristic backbone structure.
  • the backbone structure is, typically, formed by phosphodiester bonds between the sugar moiety of the nucleotide, i.e. deoxyribose, of a first and a phosphate moiety of a second, adjacent monomer.
  • the specific order of the monomers i.e.
  • DNA may be single stranded or double stranded.
  • the nucleotides of the first strand typically hybridize with the nucleotides of the second strand, e.g. by A/T-base- pairing and G/C-base-pairing.
  • a monocistronic nucleic acid may typically be a DNA or RNA, 5 particularly an mRNA that comprises only one coding sequences.
  • a coding sequence in this context is a sequence of several nucleotide triplets (codons) that can be translated into a peptide or protein
  • the coding region of the at least one mRNA of the composition may encode at least two, three, four, five, six. seven, eight and more antigens (or fragments and derivatives thereof) linked with or without an amino acid linker ID sequence, wherein said linker sequence can comprise rigid linkers, flexible linkers, cleavable linkers (e.g., self-cleaving peptides) or a combination thereaf.
  • the antigens may be identical or different or a combination thereof.
  • specific antigen and/or epitope combinations according to the paragraph "specific antigen combinations" disclosed above are particularly envisioned.
  • Particular antigen/epitope combinations can be encoded by said mRNA encoding at least two antigens as explained abovE (herein referred to as "multi-antigen-constructs/mRNA").
  • composition of the present invention may comprise a mixture of at least one monocistronic mRNA, as defined above, and/ or at least one bicistronic mRNA as defined above, and/ or at least one multieistronic mRNA, as defined above, and/ or at least one multi-antigen-constructs as defined above, and any combinations thereof.
  • specific antigen combinations according to the paragraph "specific antigen combinations" disclosedD above are particularly envisioned and may be generated using a combination of mono-, bi-, multieistronic mRNA and multi- antigen-constructs.
  • the mRNA sequence is mono-, bi-, or multieistronic, preferably as defined herein.
  • the coding sequences in a bi- or multieistronic mRNA preferably encode distinct peptides or proteins as defined5 herein or a fragment or variant thereof.
  • the coding sequences encoding two or more peptides or proteins may be separated in the bi- or multieistronic mRNA by at least one IRES (internal ribosomal entry site) sequence, as defined below.
  • IRES internal ribosomal entry site
  • the bi- or even multieistronicD mRNA may encode, for example, at least two, three, four, five, six or more (preferably different) peptides or proteins as defined herein or their fragments or variants as defined herein.
  • IRES internal ribosomal entry site
  • IRES sequences which can be used according to the invention, are those from picornavirusBS (e.g. FMDV),5 pestiviruses (CFFV), poxviruses (PV).
  • ECMV encephalomyocarditis viruses
  • FMDV foot and mouth disease viruses
  • HCV hepatitis C viruses
  • CSFV classical swine fever viruses
  • MLV mouse leukoma virus
  • SIV simian immunodeficiency viruses ar cricket paralysis viruses
  • the at least one coding region of the mRNA sequence according to the invention may encode 5 at least two, three, four, five, six, seven, eight and more peptides or proteins (or fragments and derivatives thereof) as defined herein linked with or without an amino acid linker sequence, wherein said linker sequence can comprise rigid linkers, flexible linkers, cleavable linkers (e.g., self-cleaving peptides) or a combination thereof.
  • the peptides or proteins may be identical or different or a combination thereof.
  • Particular peptide or protein combinations can be encoded by said mRNA encoding at least two peptides or proteins as explained herein (also referred to herein as "multi-antigen-constructs/mRNA").
  • T cell epitopes or parts of the proteins in the context of the present invention may comprise fragments preferably having a length of about 6 to about 2D or even more amino acids, e.g. fragments as processed and presented by MHC class I molecules, preferably having a length of about 8 to about ID amino acids, e.g.8, 9, or ID, (or even II, or I2 amino acids), or fragments as processed and presented by MHC
  • I5 class II molecules preferably having a length of about I3 or more amino acids, e.g. I3, 14, I5, IB, I7, 18, IB, 20 or even more amino acids, wherein these fragments may be selected from any part of the amino acid sequence.
  • These fragments are typically recognized by T cells in form of a complex consisting of the peptide fragment and an MHC molecule, i.e. the fragments are typically not recognized in their native form.
  • B cell epitopes are typically fragments located on the outer surface of (native) protein or peptide antigens as defined herein, preferably having 5 to I5 amino acids, more preferably having 5 to I2 amino acids, even moreD preferably having B to 3 amino acids, which may be recognized by antibodies, i.e.
  • epitopes of proteins or peptides may furthermore be selected from any of the herein mentioned variants of such proteins or peptides.
  • antigenic determinants can be conformational or discontinuous epitopes which are composed of segments of the proteins or peptides as defined herein that are discontinuous in the amino acid sequence of the proteins or peptides as defined herein but are brought together in the three-dimensional structure or continuous or linear epitopes which are composed of a single5 polypeptide chain.
  • a fragment of a sequence may typically be a shorter portion of a full-length sequence of e.g. a nucleic acid molecule or an amino acid sequence. Accordingly, a fragment, typically, consists of a sequence that is identical to the corresponding stretch within the full-length sequence.
  • a preferred fragment of a sequence in the context of the present invention,D consists of a continuous stretch of entities, such as nucleotides or amino acids corresponding to a continuous stretch of entities in the molecule the fragment is derived from, which represents at least 5%, ID , 20%, preferably at least 30%, more preferably at least 40%, more preferably at least 50%, even more preferably at least 00%, even more preferably at least 70%, and most preferably at least 80% of the total (i.e. full-length) molecule from which the fragment is derived.
  • a G/C-modified nucleic acid may typically be a nucleic acid, preferably an artificial nucleic acid molecule as defined herein, based on a modified wild type sequence comprising a preferably increased number of guanosine and/or cytosine nucleotides as compared to the wild type sequence. Such an increased number may be generated by substitution of cndons containing adenosine or thymidine nucleotides by cndons containing guanosine or cytosine nucleotides. If the enriched G/C content occurs in a coding region of DNA or RNA. it makes use of the degeneracy of the genetic code. Accordingly, the codon substitutions preferably do not alter the encoded amino acid residues, but exclusively increase the G/C content of the nucleic 5 acid molecule.
  • Gene therapy may typically be understood to mean a treatment of a patient's body or isolated Elements of a patient's body, for example isolated tissues/cells, by nucleic acids encoding a peptide or protein. It typically may comprise at least one of the steps of a) administration of a nucleic acid, preferably an artificial nucleic acid molecule as defined herein, I0 directly to the patient - by whatever administration route - or in vitro to isolated cells/tissues of the patient, which results in transfectian of the patient's cells either in vivo/ ex vivo or in vitro; b) transcription and/ or translation of the introduced nucleic acid molecule; and optionally c) re-administration of isolated, transfected cells to the patient, if the nucleic acid has not been administered directly to the patient.
  • a nucleic acid preferably an artificial nucleic acid molecule as defined herein
  • Genetic vaccination may typically be understood to be vaccination by administration of a nucleic acid molecule encoding an antigen or an immunngen or fragments thereof.
  • the nucleic acid molecule may be administered to a subject's body or to isolated cells of a subject. Upon transfection of certain cells of the body or upon transfection of the isolated cells, the antigen or immunogen may be expressed by those cells and subsequently presented to the immune system, eliciting an adaptive, i.e. antigen-specific immune response.
  • genetic vaccination typically comprises at least one of the steps ofD a) administration of a nucleic acid, preferably an artificial nucleic acid molecule as defined herein, to a subject, preferably a patient, or to isolated cells of a subject, preferably a patient, which usually results in transfection of the subject's cells either in vivo or in vitro; b) transcription and/or translation of the introduced nucleic acid molecule; and optionally c) re-administration of isolated, transfected cells to the subject, preferably the patient, if the nucleic acid has not been administered directly to the patient.
  • a nucleic acid preferably an artificial nucleic acid molecule as defined herein
  • Genotype, genotype of a virus The terms "genotype” or “genotype of a virus” have to be understood as the genetic constitution of an individual or a group or class of organisms having the same genetically consistent structure. Genotyping means determining differences in the genetic of an individual. In the context of the invention, Noruvirus genotype has to be understood as a Noro virus having the same genetically consistent structure.
  • Heterologous sequence Two sequences are typically understood to be “heterologous” if they are not derivable from the same gene or in the same allele. I.e., although heterologous sequences may be derivable from the same organism, they naturally (in nature) do not occur in the same nucleic acid molecule, such as in the same mRNA.
  • H p fTiDioq of a nucleic acid sequence The term "homolog" of a nucleic acid sequence refers to sequences of other species than the particular sequence. It is particularly preferred that the nucleic acid sequence is of human origin and therefore it is preferred that the homolog is a homolog of a human nucleic acid sequence.
  • Humoral immunity refers typically to antibody production and optionally to accessory processes accompanying antibody production.
  • a humoral immune respanse may be typically characterized, e.g., by Th2 activation and cytokine production, germinal center formation and isotype switching, affinity maturation and memory cell generation.
  • Humoral immunity also typically may refer to the effector functions of antibodies, which include pathogen and toxin neutralization, classical complement activation, and opsonin promotion of phagocytosis and pathogen elimination.
  • an immunogen may be typically understood to be a compound that is able to stimulate an immune respanse.
  • an immunogen is a peptide, polypeptide, or protein.
  • an immunogen in the sense of the present invention is the product of translation of a provided nucleic acid molecule, preferably an artificial nucleic acid molecule as defined herein.
  • an immunogen elicits at least an adaptive immune I5 response.
  • an immunostimulatory composition may be typically understood to be a composition containing at least one component which is able to induce an immune response or from which a component which is able to induce an immune response is derivable. Such immune response may be preferably an innate immune response0 or a combination of an adaptive and an innate immune response.
  • an immunostimulatory composition in the context of the invention contains at least one artificial nucleic acid molecule, more preferably an HA, for example an mRNA molecule.
  • the immunostimulatory component such as the m NA may be complexed with a suitable carrier.
  • the immunostimulatory composition may comprise an mRNA/carrier-camplex.
  • the immunostimulatory composition may comprise an adjuvant and/ or a suitable vehicle for the immunostimulatory component, such as the mRNA.
  • Immune response An immune response may typically be a specific reaction of the adaptive immune system to a particular antigen (so called specific or adaptive immune response) or an unspecific reaction of the innate immune system (so called unspecific or innate immune response), or a combination thereof.
  • the immune system may protect organisms from infection. If a pathogen succeeds in passing a physical barrier of an organism and enters this organism, the innate immune system provides an immediate, but non-specific response. If pathogens evade this innate response, vertebrates possess a second layer of protection, the adaptive immune system. Here, the immune system adapts its response during an infection to improve its recognition of the pathogen.
  • the immune system comprises the innate and the adaptive immune system. Each of these two parts typically contains so called humoral and cellular components.
  • Immunostimulatory RNA in the context of the invention may typically be an RNA that is able 5 to induce an innate immune response. It usually does not have a coding region and thus does not provide a peptide-antigen or immunogen but elicits an immune response e.g. by binding to a specific kind of Toll-like-receptor (TLR) or other suitable receptors.
  • TLR Toll-like-receptor
  • mRNAs having a coding region and coding for a peptide/protein may induce an innate immune response and, thus, may be immunostimulatory RNAs.
  • the innate immune system also known as non-specific (or unspecific)
  • the immune system typically comprises the cells and mechanisms that defend the host from infection by other organisms in a non ⁇ specific manner. This means that the cells of the innate system may recognize and respond to pathogens in a generic way, but unlike the adaptive immune system, it does not confer long-lasting or protective immunity to the host.
  • the innate immune system may be. e.g., activated by ligands of Toll-like receptors (TLRs) or other auxiliary substances such as lipopnlysaccharides, TNF-
  • the pharmaceutical composition according to the present invention may comprise one or more such substances.
  • a response of the innate immune system includes recruiting immune cells to sites of infection, through the production of chemical factors, including specialized chemical mediators, called cytokines; activation of the complement cascade; identification and removal of foreign substances present in organs, tissues, the blood and lymph, by specialized white blood cells;5 activation of the adaptive immune system; and/ or acting as a physical and chemical barrier to infectious agents.
  • Jet injection refers to a needle-free injection method, wherein a fluid containing at least one inventive nucleic acid seguence (e.g., RNA, DNA, mRNA) and, optionally, further suitable excipients is forced through an orifice, thus generating an ultra-fine liquid stream of high pressure that is capable of penetrating mammaliao skin and, depending on theD injection settings, subcutaneous tissue or muscle tissue.
  • the liquid stream forms a hole in the skin, through which the liquid stream is pushed into the target tissue.
  • jet injection is used for intradermal, subcutaneous or intramuscular injection of the mRNA sequence according to the invention.
  • jet injection is used for intramuscular injection of the mRNA sequence according to the invention.
  • jet injection is used for intradermal injection of the mRNA sequence according to the invention.
  • a monovalent vaccine also called univalent vaccine, is designed against a single antigen for a single organism.
  • the term "monovalent vaccine” includes the immunization against a single valence.
  • a monovalent Norovirus vaccine would comprise a vaccine comprising an artificial nucleic acid encoding one single antigenic peptide or protein derived from one specific Norovirus strain.
  • Nucleic acid molecule A nucleic acid molecule, an artificial nucleic acid, or nucleic acid is a molecule comprising, preferably consisting of nucleic acid components.
  • the terms nucleic acid molecule, artificial nucleic acid, or nucleic acid preferably refer to DNA or RNA molecules and vice versa. It is preferably used synonymous with the term "palynucleotide".
  • a nucleic acid molecule is a polymer comprising or consisting of nucleotide monomers, which are covalently linked to each other by phosphodiester-bonds of a sugar/phosphate-backbone.
  • nucleic acid molecule also encompasses modified nucleic acid molecules, such as base-modified, sugar-modified or backbone-modified etc. It encompasses any type of DNA or RNA molecules.
  • Nucleic acid sequence/amino acid The sequence of a nucleic acid molecule is typically understood to be the particular and individual order, i.e. the succession of its nucleotides.
  • sequence of a protein or peptide is typically understood to be the order, i.e. the succession of its amino acids.
  • a peptide or polypeptide is typically a polymer of amino acid monomers, linked by peptide bonds. It typically contains less than 5D monomer units. Nevertheless, the term peptide is not a disclaimer for molecules having more than 50 monomer units. Long peptides are also called polypeptides, typically having between 50 and 600 monomeric units.
  • the term "polypeptide” as used herein, however, is typically not limited by the length of the molecule it refers to. In the context of the present invention, the term “polypeptide” may also be used with respect to peptides comprising less than 50 (e.g. I0) amino acids or peptides comprising even more than GOO amino acids. Also, the terms “polypeptide”, “peptide”, and “protein” are used interchangeably herein to refer to polymers of amino acids of any length.
  • a pharmaceutically effective amount in the context of the invention is typically understood to be an amount that is sufficient to induce a pharmaceutical effect, such as an immune response, altering a pathological level of an expressed peptide or protein, or substituting a lacking gene product, e.g., in case of a pathological situation.
  • a protein typically comprises one or more peptides or polypeptides.
  • a protein is typically folded into S-dimensional form, which may be required for the protein to exert its biological function.
  • a poly(A) sequence also called poly(A) tail or 3'-poly(A) tail, is typically understood to be a sequence of adenosine nucleotides, e.g., of up to about 4D0 adenosine nucleotides, e.g. from about 2D to about 4DD, preferably from about 50 to about 40D, more preferably from about 50 to about 300, even more preferably from about 50 to about 250, most preferably from about B0 to about 250 adenosine nucleotides.
  • a poly(A) sequence is typically located at the 3'-end of an mRNA.
  • a poly(A) sequence may be located within an m NA or any other nucleic acid molecule, such as, e.g., in a vector, for example, in a vector serving as template for the generation of an RNA, preferably an mRNA, e.g., by transcription of the vector.
  • poly(A) sequences, or poly(A) tails may be generated in vitro by enzymatic polyadenylation of the RNA, e.g. using Pnly(A)polymerases (PAP) derived from E.coli or yeast.
  • PAP Pnly(A)polymerases
  • polyadenylation of RNA can be achieved by using 5 immobilized PAP eozymes e.g. in a polyadenylation reactor (WD 2DIB/I7427I).
  • a poly(C) sequence is typically a long sequence of cytosine nucleotides, typically about ID to about 2D0 cytosine nucleotides, preferably about ID to about I00 cytosine nucleotides, more preferably about ID to about 7D cytosine nucleotides or even more, preferably about 2D to about 50, or even about 2D to about 3D cytosine nucleotides.
  • a poly(C) sequence may ID preferably be located 3' of the coding sequence comprised by a nucleic acid.
  • Polyadenylation is typically understood to be the addition of a poly(A) sequence to a nucleic acid molecule, such as an RNA molecule, e.g. to a premature mRNA. Polyadenylation may be induced by a so called polyadenylation signal. This signal is preferably located within a stretch of nucleotides at the 3'-end of a nucleic acid molecule, such as an RNA molecule, to be I5 polyadenylated.
  • a polyadenylation signal typically comprises a hexamer consisting of adenine and uracil/thymine nucleotides, preferably the hexamer sequence AAUAAA.
  • Polyadenylation typically occurs during processing of a pre-mRNA (also called premature-mRNA).
  • RNA maturation comprises the step of polyadenylation.
  • Polyvalent/polyvalent vaccine A polyvalent vaccine, called also multivalent vaccine, containing antigens from more than one strain of a virus, or different antigens of the same virus, or any combination thereof.
  • the term "polyvalent vaccine” describes that this vaccine has more than one valence.
  • a polyvalent Norovirus vaccine would comprise a vaccine camprising an artificial nucleic acid encoding antigenic peptides or proteins derived from several different Norovirus strains or comprising artificial nucleic acid encoding different antigens from the same Norovirus strain, or a combination thereof.5
  • a polyvalent Norovirus vaccine comprises 2, 3, 4, 5, B, 7, 8, 9, ID, II, I2, 13, 14, 15, IB, I7, 18, 19, 2D, 21, 22, 23, 24, 25, 2B, 27, 28, 2D, 30, 31, 32, 33, 34, 35, 3B, 37, 38, 39, 0, 142, 43, 4, 45, 4B, 47.48, 49, 50, 51, 52, 53, 54, 55, 5B, 57, 58, 59, BO, Bl, B2, B3, B4, B5, BB, B7, B8, B9, 70, 71, 72, 3, 74, 75, 7B, 77, 78, 79, 80, 81, 82, 83
  • restriction site also termed restriction enzyme recognition site, is a nucleotide sequence recognized by a restriction enzyme.
  • a restriction site is typically a short, preferably palindromic nucleotide sequence, e.g. a sequence comprising 4 to 8 nucleotides.
  • a restriction site is preferably specifically recognized by a restriction enzyme.
  • the restriction enzyme typically cleaves a nucleotide sequence comprising a restriction site at this site.
  • the restrictioo enzyme typically cuts both strands of the nucleotide sequence.
  • NA, mRNA RNA is the usual abbreviation for ribonucleic-acid.
  • nucleic acid molecule i.e. a polymer consisting of nucleotides.
  • nucleotides are usually adenosine-monophosphate, uridine-monophosphate, guanosine-monophosphate and cytidine-monophosphate monomers which are connected to each other along a so-called backbone.
  • the backbone is formed by phosphodiester bonds between the sugar, i.e. ribose, of a first and a phosphate moiety of a second, adjacent monomer.
  • RNA-sequence 5 specific succession of the monomers is called the RNA-sequence.
  • RNA may be obtainable by transcription of a DNA- sequence, e.g., inside a cell. In eukaryotic cells, transcription is typically performed inside the nucleus or the mitochondria. Typically, transcription of DNA usually results in the so-called premature RNA which has to be processed into so-called messenger-RNA, usually abbreviated as mRNA. Processing of the premature RNA, e.g. in eukaryotic organisms, comprises a variety of different posttranscriptional-modifications such as splicing. 5'-capping, polyadenylation, export from the nucleus or
  • RNA the mitochondria and the like.
  • the sum of these processes is also called maturation of RNA.
  • the mature messenger RNA usually provides the nucleotide sequence that may be translated into an amino-acid sequence of a particular peptide or protein.
  • a mature mRNA comprises a 5'-cap, a 5'-UTR, a coding region, a 3'-UTR and a poly(A) sequence.
  • I5 Stabilized nucleic acid preferably mRNA:
  • a stabilized nucleic acid, preferably mRNA typically, exhibits a modification increasing resistance to in vivo degradation (e.g. degradation by an BXO- or endo-nuclease) and/or ex vivo degradation (e.g. by the manufacturing process prior to vaccine administration, e.g. in the course of the preparation of the vaccine solution to be administered).
  • Stabilization of RNA can, e.g., be achieved by providing a 5'-cap-Structure, a Poly-A-Tail, or any other UTR- modification. It can also be achieved by chemical modification or modification of the G/C content of the nucleic acid or other0 types of sequence optimization.
  • Various other methods are known in the art and conceivable in the context of the invention.
  • Sequence identity Two or more sequences are identical if they exhibit the same length and order of nucleotides or amino acids.
  • the percentage of identity typically describes the extent, to which two sequences are identical, i.e. it typically describes the percentage of nucleotides that correspond in their sequence position with identical nucleotides of a reference sequence.
  • the sequences to be compared are considered to exhibit the same length, i.e. the length of the longest sequence of the sequences to be compared. This means that a first sequence consisting of 8 nucleotides is 80% identical to a second sequence consisting of ID oucleotides comprising the first sequence.
  • identity of sequences preferably relates to the percentage of nucleotides of a seqoence which have the same position in two or more sequences having the same length. Therefore, e.g. a position of a first sequence may be compared with theD corresponding position of the second sequence. If a position in the first sequence is occupied by the same component (residue) as is the case at a position in the second sequence, the two sequences are identical at this position. If this is not the case, the sequences differ at this position. If insertions occur in the second sequence in comparison to the first sequence, gaps can be inserted into the first sequence to allow a further alignment.
  • deletions occur io the second sequence in comparison to the first sequence, gaps can be inserted into the second sequence to allow a further alignment.
  • the percentage to which two seqoences5 are identical is then a function of the number of identical positions divided by the total number of positions including those positions which are only occupied in ooe sequence.
  • the percentage to which two sequences are identical can be determined using a mathematical algorithm.
  • a preferred, but not limiting, example of a mathematical algorithm which can be used is the algorithm of Karlin et al. ((903), PNAS USA, 90:5873-5877 or Altschul et al. (1997), Nucleic Acids Res., 25:3399-3402. Such an algorithm is integrated in the BLAST program. Sequences which are identical to the sequences of the present invention to a certain extent can be identified by this program.
  • Serotype, serotype of a virus is a group of viruses classified together based on their antigens on the surface of the virus, allowing the epidemiologic classification of organisms to the sub-species level.
  • strain of a virus A strain or a strain of a virus is a group of viruses that are genetically distinct from other groups of the ID same species.
  • the strain that is defined by a genetic variant is also defined as a "subtype".
  • Transfectian refers to the introduction of nucleic acid molecules, such as DNA or NA (e.g. mRNA) molecules, into cells, preferably into eukaryotic cells.
  • nucleic acid molecules such as DNA or NA (e.g. mRNA) molecules
  • transfection encompasses any method known to the skilled person for introducing nucleic acid molecules into cells, preferably into eukaryotic 15 cells, such as into mammalian cells. Such methods encompass, for example, electroporation, lipofection, e.g.
  • D Vaccine A vaccine is typically understood tn be a prophylactic or therapeutic material providing at least one antigen, preferably an immunogen.
  • the antigen or immunogen may be derived from any material that is suitable for vaccination.
  • the antigen or immunogen may be derived from a pathogen, such as from bacteria or virus particles etc., or from a tumor or cancerous tissue.
  • the antigen or immunogen stimulates the body's adaptive immune system to provide an adaptive immune response.
  • Vector refers to a nucleic acid molecule, preferably to an artificial nucleic acid molecule.
  • a vector in the context of the present invention is suitable for incorporating or harboring a desired nucleic acid sequence, such as a nucleic acid sequence comprising a coding region.
  • Such vectors may be storage vectors, expression vectors, cloning vectors, transfer vectors etc.
  • a storage vector is a vector which allows the convenient storage of a nucleic acid molecule, for example, of an mRNAD molecule.
  • the vector may comprise a sequence corresponding, e.g., to a desired mRNA sequence or a part thereof, such as a sequence corresponding to the coding region and the 3'-UTR and/or the 5'-UTR of an mRNA.
  • An expression vector may be used for production of expression products such as RNA, e.g. mRNA, or peptides, polypeptides or proteins.
  • an expression vector may comprise sequences needed for transcription of a sequence stretch of the vector, such as a promoter sequence, e.g. an RNA polymerase promoter sequence.
  • a cloning vector is typically a vector that contains a cloning site, which may be used5 to incorporate nucleic acid sequences into the vector.
  • a cloning vector may be, e.g., a plasmid vector or a bacteriophage vector.
  • a transfer vector may be a vector which is suitable for transferring nucleic acid molecules into cells or organisms, for example, viral vectors.
  • a vector in the context of the present invention may be, e.g., an RNA vector or a ONA vector.
  • a vector is a DNA molecule.
  • a vector in the sense of the present application comprises a cloning site, a selection marker, such as an antibiotic resistance factor, and a sequence suitable for multiplication of the vector, such as an origin of replication.
  • a vector in the context of the present application is a plasmid vector.
  • RNA in vitro transcription or “in vitro transcription” relate to a process wherein RNA is synthesized in a cell-free system (in vitro).
  • DNA particularly plasmid DNA
  • RNA is used as template for the generation of RNA transcripts.
  • RNA may be obtained by DNA-depeodent in vitro transcription of an appropriate DNA template, which according to the present invention is preferably a linearized plasmid DNA template.
  • the promoter for controlling in vitro transcription can be any promoter for any DNA-dependent RNA polymerase.
  • DNA-dependent RNA polymerases are the T7, T3, and SPG RNA polymerases.
  • a DNA template for in vitro RNA transcription may be obtained by cloning of a nucleic acid, in particular cDNA corresponding to the respective RNA to be in vitro transcribed, and introducing it into an appropriate vector for in vitro transcription, for example into plasmid DNA.
  • the DNA template is linearized with a suitable restriction enzyme, before it is transcribed in vitro.
  • the cDNA may be obtained by reverse transcription of mRNA or chemical synthesis.
  • the DNA template for in vitro RNA synthesis may also be obtained by gene synthesis.
  • RNA polymerase such as bacteriophage-encoded RNA polymerases
  • NTPs ribonucleoside triphosphates
  • a cap analogue as defined above e.g. m7G(5')ppp(5')G (m7G)
  • fraction of NTPs optimized to the RNA sequence accordinging to WD/2DI5/I88933;
  • RNA-dependent RNA polymerase capable of binding to the promoter sequence within the linearized DNA template (e.g.
  • T7, T3 or SPG RNA polymerase
  • RNase ribonuclease
  • G optionally a pyrophosphatase to degrade pyrophosphate, which may inhibit transcription;
  • MgCI2 which supplies Mg2+ ions as a co-factor for the polymerase
  • a buffer to maintain a suitable pH value which can also contain antioxidants (e.g. DTT), and/or polyamines such as spermidine at optimal concentrations.
  • antioxidants e.g. DTT
  • polyamines such as spermidine
  • a vehicle is typically understood to be a material that is suitable for storing, transporting, and/or administering a compound, such as a pharmaceutically active compound.
  • a compound such as a pharmaceutically active compound.
  • it may be a physiologically acceptable liquid which is suitable for storing, transporting, and/or administering a pharmaceutically active compound.
  • Different species/species defines a monophyletic group of viruses whose properties can be distinguished from those of other species by multiple criteria (Adams et al., 2DI3, Arch Virol I58: 2633-9).
  • Nnrovirus or Noroviruses as used in the present invention means that a Norovirus from another species, strain 5 or serotype is used or that the properties of the Noroviruses which are used or utilized can be distinguished from those of the other Norovirus by multiple criteria.
  • 3'-DTR ⁇ '-untranslated region
  • 3'-DTR refers to a part of the artificial nucleic acid molecule, which is located 3' (i.e. "downstream") of a coding region and which is not translated into protein.
  • a 3'-UTR is the part of an
  • the term 3'-DTR may also comprise elements, which are not encoded in the template, from which an RNA is transcribed, but which are added after transcription during maturation, e.g. a poly(A) sequence.
  • a 3'-UTR of the mRNA is not translated into an amino acid sequence.
  • the 3'-UTR sequence is generally encoded by the gene which is transcribed into the respective mRNA during the gene expression process. The genomic sequence is first transcribed into pre-
  • the pre-mature mRNA is then further processed into mature mRNA in a maturation process.
  • This maturation process comprises the steps of 5'capping, splicing the pre-mature mRNA to excize optional introns and modifications of the 3'-end, such as polyadenylation of the 3'-end of the pre-mature mRNA and optional endo-/ or exonuclease cleavages etc.
  • a 3'-DTR corresponds to the sequence of a mature mRNA which is located between the stop codon of the protein coding region, preferably immediately 3' to the stop codon of the proteinD coding region, and the poly(A) sequence of the mRNA.
  • the term "corresponds to” means that the 3'-DTR sequence may be an RNA sequence, such as in the mRNA sequence used for defining the 3'-DTR sequence, or a DNA sequence which corresponds to such RNA sequence.
  • the term "a 3'-DTR of a gene" is the sequence which corresponds to the 3'-DTR of the mature mRNA derived from this gene, i.e.
  • 3'-DTR of a gene encompasses the DNA sequence and the RNA sequence (both sense and antisense5 strand and both mature and immature) of the 3'-UTR.
  • the 3'-UTRs have a length of more than 2D, 3D, 40 or 50 nucleotides.
  • 5'-untranslated region S'-OTR: Generally, the term “5'-UTR” refers to a part of the artificial nucleic acid molecule, which is located 5' (i.e. "upstream") of a coding region and which is not translated into protein.
  • a 5'-DTR is typically understood to be aD particular section of messenger RNA (mRNA), which is located 5' of the coding region of the mRNA.
  • mRNA messenger RNA
  • the 5'-UTR starts with the transcriptional start site and ends one nucleotide before the start codon of the coding region.
  • the 5'-UTRs have a length of more than 2D, 3D, 40 or 50 nucleotides.
  • the 5'-UTR may comprise elements for controlling gene expression, also called regulatory elements. Such regulatory elements may be, for example, ribosomal binding sites.
  • the 5'-UTR may be post transcriptionally modified, for example by addition of a 5'-CAP.
  • a 5'-DTR of the mRNA is not translated into an amino acid5 sequence.
  • the 5'-DTR sequence is generally encoded by the gene which is transcribed into the respective mRNA during the gene expression process.
  • the genomic sequence is first transcribed into pre-mature mRNA, which comprises optional introns.
  • the pre-mature mRNA is then further processed into mature mRNA in a maturation process.
  • This maturation process comprises the steps of 5'capping, splicing the pre-mature mRNA to excize optional intrnns and modifications of the S'-end. such as polyadenylation of the S'-end of the pre-mature mRNA and optional endoV or exonuclease cleavages etc..
  • a 5'-UTR corresponds to the sequence of a mature mRNA which is located between the start codon and, for
  • the 5'-UTR corresponds to the sequence which extends from a nucleotide located 3' to the 5'- cap, more preferably from the nucleotide located immediately 3' to the 5'-eap, to a nucleotide located 5' tn the start codon of the protein coding region, preferably to the nucleotide located immediately 5' to the start codon of the protein coding region.
  • the nucleotide located immediately 3' to the 5'-cap of a mature mRNA typically corresponds to the transcriptional start site.
  • the term "corresponds to" means that the 5'-UTR sequence may be an RNA sequence, such as in the mRNA sequence used for defining
  • a 5'-UTR of a gene is the sequence which corresponds to the 5'-DTR of the mature mRNA derived from this gene, i.e. the mRNA obtained by transcription of the gene and maturation of the pre-mature mRNA.
  • the term "5'-UTR of a gene” encompasses the DNA sequence and the RNA sequence (both sense and antisense strand and both mature and immature) of the 5'-UTR.
  • TDP I5 5'-Terminal Oligapyrimidine Tract
  • the 5'-terminal oligopyrimidine tract (TDP) is typically a stretch of pyrimidine nucleotides located in the 5'-terminal region of a nucleic acid molecule, such as the 5'-terminal region of certain mRNA molecules or the 5'-terminal region of a functional entity, e.g. the transcribed region, of certain genes.
  • the sequence starts with a cytidine, which usually corresponds to the transcriptional start site, and is followed by a stretch of usually about 3 to 3D pyrimidine nucleotides.
  • the TDP may comprise 3, 4, 5, 6, 7. 8, B, ID, II, I2, 13, 14, I5, IB.
  • TDP mRNA messenger RNA that contains a 5'-terminal oligopyrimidine tract
  • TOP genes genes that provide such messenger RNAs are referred tn as TOP genes.
  • TOP sequences have, for example, been found in genes and mRNAs encoding peptide elongation factors and ribosomal proteins.
  • a TOP motif is a nucleic acid sequence which corresponds to a 5'TOP as defined above.
  • a TDP motif in the context of the present invention is preferably a stretch of pyrimidine nucleotides having a length of 3-30 nucleotides.
  • the TOP-motif consists of at least 3 pyrimidine nucleotides, preferably at least 4 pyrimidine nucleotides, preferably at least 5 pyrimidine nucleotides, more preferably at least G nucleotides, more preferably at least 7 nucleotides, most preferably at least 8 pyrimidine nucleotides, wherein the stretch of pyrimidine nucleotides preferably starts0 at its 5'-end with a cytosine nucleotide.
  • the TOP-motif preferably starts at its S'-end with the transcriptional start site and ends one nucleotide 5' to the first purin residue in said gene or mRNA.
  • a TOP motif in the sense of the present invention is preferably located at the 5'-end of a sequence which represents a 5'-UTR or at the 5'-end of a sequence which codes for a 5'-DTR.
  • TDP motif a stretch of 3 or more pyrimidine nucleotides is called "TDP motif" in the sense of the present invention if this stretch is located at the 5'-end of a respective sequence, such as the artificial nucleic acid molecule,5 the 5'-0TR element of the artificial nucleic acid molecule, or the nucleic acid sequence which is derived from the 5'-0TR of a TOP gene as described herein.
  • a stretch of 3 or more pyrimidine nucleotides which is not located at the 5'-end of a 5'-UTR or a 5'-UTR element but anywhere within a 5'-UTR or a 5'-UTR element, is preferably not referred to as "TDP motif".
  • TDP genes are typically characterised by the presence of a 5'-terminal oligopyrimidine tract. Furthermore, most TOP genes are characterized by a growth-associated translational regulation. However, also TOP genes with a tissue specific translational regulation are known. As defined above, the 5'-0TR of a TOP gene corresponds to the sequence of a 5'-DTR of a mature mRNA derived from a TDP gene, which preferably extends from the nucleotide located 3' to the 5'-cap to the nucleotide located 5' to the start codon. A 5'-UTR of a TOP gene typically does not comprise any start codons, preferably no upstream AUGs (uAUGs) or upstream coding regions (uDRFs).
  • uAUGs upstream AUGs
  • uDRFs upstream coding regions
  • upstream AUGs and upstream coding regions are typically understood to be AOGs and coding regions that occur 5' of the start codon (AOG) of the coding region that should be translated.
  • the 5'-UTRs of TDP genes are generally rather short.
  • the lengths of 5'-DTRs of TOP genes may vary between ZD nucleotides up to 5DD nucleotides, and are typically less than about 2D0 nucleotides, preferably less than about I5D nucleotides, more preferably less than about 100 nucleotides.
  • Exemplary 5'-0TRs of TDP genes in the sense of the present invention are the nucleic acid sequences extending from the nucleotide at position 5 to the nucleotide located immediately 5' to the start codon (e.g. the ATG) in the sequences according to SED ID NQs: I-I3G3 of the patent application WO 2013/143700. whose disclosure is incorporated herewith by reference.
  • a particularly preferred fragment of a 5'-0TR of a TOP gene is a 5'-DTR of a TDP gene lacking the 5'TOP motif.
  • the terms "5'-UTR of a TOP gene" or "5'-T0P UTR" preferably refer to the 5'-UTR of a naturally occurring TOP gene.
  • Orthologues and paralogues encompass evolutionary concepts used to describe the ancestral relationships of genes. Paralogues are genes within the same species that have originated through duplication of an ancestral gene; orthologues are genes from different organisms that have originated through speciation, and are also derived from a common ancestral gene.
  • an orthologue and/or a paralague of a Norovirus nucleic acid sequence of the invention preferably refers to a sequence having in increasing order of preference at least 50%, 51%, 52%, 53%, 54%. 55%, 56%. 57%, 58%, 59%.
  • an orthologue and/or a paralague of a Norovirus amino acid sequence of the invention refers preferably to a sequence having in increasing order of preference at least 50%. 51%. 52%, 53%, 54%, 55%, 56%, 57%. 58%. 59%. G0%. 61%. 62%, 63%. 64%, 65%, 66%, 67%, 68%, 69%. 70%. 71%, 72%, 73%, 74%, 75%, 76%. 77%, 78%, 79%, 80%. 81%. 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89% 90%. 91%, 92%, 93%. 94%, 95%, 96%, 97%, 98%, or 99% or more sequence identity to the sequence as represented by SEQ 10 NOs: 1-4410.
  • Hybridization/Homoloqy Nucleic acid molecules which are advantageously for the process according to the invention can be isolated based on their homology to the nucleic acid molecules or a complement sequence of the nucleic acid molecules disclosed herein using the sequences or part thereof as hybridization probe and following standard hybridization techniques under stringent hybridization conditions.
  • nucleic acid molecules of at least 15, 2D, 25, 3D, 35, 4D, 5D, BD or more nucleotides preferably of at least 15, 20 or 25 nucleotides in length which hybridize under stringent conditions with the above-described nucleic acid molecules, in particular with those which encompass a nucleotide sequence of the nucleic acid molecule used in the invention or encoding a protein used in the invention or of the nucleic acid molecule of the invention.
  • Nucleic acid molecules with 3D, 5D, IOD, 25D or more nucleotides may also be used.
  • nucleic acid molecules that are homologous to the nucleic acid molecules described above and that are derivatives of said nucleic acid molecules are, for example, variations of said nucleic acid molecules which represent modifications having the same biological function, in particular encoding proteins with the same or substantially the same biological function. They may be naturally occurring variations, such as sequences from other species, strains, or mutations. These mutations may occur
  • allelic variations naturally or may be obtained by mutagenesis techniques.
  • allelic variations may be naturally occurring allelic variants as well as synthetically produced or genetically engineered variants.
  • Structurally equivalents can, for example, be identified by testing the binding of said polypeptide to antibodies or computer based predictions.
  • hybridizing it is meant that such nucleic acid molecules hybridize under conventional hybridization conditions, preferably under stringent conditions such as described by, e.g., Sambrook (Molecular Cloning; A Laboratory Manual, 2nd Edition, Cold Spring 15 Harbor Laboratory Press, Cold Spring Harbor, NY (1989)) or in Current Protocols in Molecular Biology, John Wiley B Sons, N. Y.
  • DNA as well as NA molecules of the nucleic acid of the invention can be used as probes.
  • Northern blot assays as well as Southern blot assays can be performed.
  • the Northern blot assay advantageously provides further information about the expressed gene product: e.g. expression pattern,D occurrence of processing steps, like splicing and capping, etc.
  • the Southern blot assay provides additional information about the chromosomal localization and organization of the gene encoding the nucleic acid molecule of the invention.
  • SSC sodium chloride/sodium citrate
  • 0.1% SDS 0.1% SDS at 5D to B5°C, for example at 50 a C, 55 D C or BD°C.
  • these hybridization conditions differ as a function of the type of the nucleic acid5 and, for example when organic solvents are present, with regard to the temperature and concentration of the buffer.
  • the temperature under "standard hybridization conditions” differs for example as a function of the type of the nucleic acid between 42°C and 58 D C, preferably between 45 D C and 5D D C in an aqueous buffer with a concentration of D.I x D.5 x, I x, 2x, 3x, 4x or 5 x SSC (pH 7.2). If organic solvent(s) is/are present in the abovementioned buffer, for example 50% formamide, the temperature under standard conditions is approximately 40 a C, 42 D C or 45 D C.
  • the hybridization conditions for DNA:DNA hybrids are preferably0 for example 0.1 x SSC and 20 D C, 25 D C, 3D D C, 35 D C, 40°C or 45 D C, preferably between 30°C and 45 n C.
  • the hybridization conditions for DNA:RNA hybrids are preferably for example 0.1 x SSC and 3D D C, 35°C. 4D°C, 45°C, 5D D C or 55°C, preferably between 45 D C and 55 D C.
  • a further example of one such stringent hybridization condition is hybridization at 4XSSC at G5 D G, followed by a washing in O.IXSSC at G5°C for one hour.
  • an exemplary stringent hybridization condition is in 50% formamide, 4XSSC at 42 D C.
  • the conditions during the wash step can be selected from the range of conditions delimited by low-stringency conditions (approximately 2X SSC at 50°C) and high-stringency conditions (approximately 0.2X SSC at 50 D C, preferably at B5 D C) (20X SSC: D.3M sodium citrate, 3M NaCI, pH 7.0).
  • the temperature during the wash step can be raised from low-stringency conditions at room temperature, approximately 22°C, to higher-stringency conditions at approximately G5°C.
  • Both of the parameters salt concentration and temperature can be varied simultaneously, or else one of the two parameters can be kept constant while only the other is varied.
  • Denaturants for example formamide or SDS, may also be employed during the hybridization. In the presence of 50% formamide, hybridization is preferably effected at 42 D C. Relevant factors like i) length of treatment, ii) salt conditions, iii) detergent conditions, iv) competitor DNAs, v) temperature and vi) probe selection can be combined case by case so that not all possibilities can be mentioned herein.
  • Hybridization conditions can be selected, for example, from the following conditions:
  • Wash steps can be selected, for example, from the following conditions:
  • a Southern blot analysis of total DNA could be probed with a nucleic acid molecule of the present invention and washed at low stringency (55 D C in 2xSSPE, 0,1% SDS).
  • a further example of such low-stringent hybridization conditions is 4XSSC at 5D°C or hybridization with 3D to 40% formamide at 42 D C.
  • Such molecules comprise those which are fragments, analogues or derivatives of the polypeptide of the invention or used in the methods of the invention and differ, for example, by way of amino acid and/or nucleotide deletion(s), insertion(s), substitution (s), addition(s) and/or recombination (s) or any other modification(s) known in the art either alone or in combination from the above-described amino acid sequences or their underlying nuclentide sequence(s).
  • Hybridization should advantageously be carried out with fragments of at least 5, 10, 15, 2D, 25, 30, 35 or 4D bp, advantageously at least 5D, BD, 7D or 80 bp, preferably at least 00, 100 or 110 bp. Most preferably are fragments of at least 15, 2D, 25 or 30 bp. Preferably are also hybridizations with at least IDD bp Dr 200, very especially preferably at least 400 bp in length. In an especially preferred embodiment, the hybridization should be carried out with the entire nucleic acid sequence with conditions described above.
  • hybridizes under stringent conditions is defined above.
  • the term “hybridizes under stringent conditions” is intended to describe conditions far hybridization and washing under which nucleotide sequences at least 30%, 40%, 50% or 65% identical to each other typically remain hybridized to each other.
  • the conditions are such that sequences at least about 70%, more preferably at least about 75% or 80%. and even more preferably at least about 85%, B0% or 35% or more identical to each other typically remain hybridized to each other,
  • the sequences are written one underneath the other for an optimal comparison (for example gaps may be inserted into the sequence of a protein or of a nucleic acid in order to generate an optimal alignment with the other protein or the other nucleic acid).
  • amino acid residues or nucleic acid molecules at the corresponding amino acid positions or nucleotide positions are then compared. If a position in one sequence is occupied by the same amino acid residue or the same nucleic acid molecule as the corresponding position in the other sequence, the molecules are homologous at this position (i.e. amino acid or nucleic acid "homology” as used in the present context corresponds to amino acid or nucleic acid "identity”.
  • Results of high quality are reached by using the algorithm of Needleman and Wunsch or Smith and Waterman. Therefore programs based on said algorithms are preferred.
  • the comparisons of sequences can be done with the program PileUp ( J. Mol. Evolution., 25, 351 (1987), Higgins et al., CABI0S 5, 151 (1989)) or preferably with the programs "Gap” and “Needle”, which are both based on the algorithms of Needleman and Wunsch (J. Mol. Biol. 48; 443 (1970)), and "BestFit", which is based on the algorithm of Smith and Waterman (Adv. Appl. Math. 2; 482 (1981 )).
  • Gap and “BestFit” are part of the GCG software-package (Genetics Computer Group, 575 Science Drive, Madison, Wisconsin, USA 53711 (1991); Altschul et al., (Nucleic Acids Res. 25, 3389 (1937)), "Needle” is part of the European Molecular Biology Dpen Software Suite (EMBOSS) (Trends in Genetics IB (B), 27B (200D)). Therefore preferably the calculations to determine the percentages of sequence homology are done with the programs "Gap” or “Needle” over the whole range of the sequences.
  • EMBOSS European Molecular Biology Dpen Software Suite
  • sequence SED ID NO: 4411 For example a sequence, which has 80% homology with sequence SED ID NO: 4411 at the nucleic acid level is understood as meaning a sequence which, upon comparison with the sequence SED ID ND: 4411 by the above program "Needle" with the above parameter set, has a 80% identity.
  • Homology between two polypeptides is understood as meaning the identity of the amino acid sequence over in each case the entire sequence length which is calculated by comparison with the aid of the above program "Needle” using Matrix: EBL0SUMG2, Gap_penalty: 8.D, Extend_penalty: 2.D.
  • sequence which has a 80% homology with sequence SEQ ID NO: I at the protein level is understood as meaning a sequence which, upon comparison with the sequence SEQ ID NO: I by the above program "Needle" with the above parameter set, has a 80% identity.
  • the invention relates to an artificial nucleic acid comprising or consisting of at least one coding region encoding at least one polypeptide derived from a Norovirus, and/or a fragment or variant thereof.
  • the artificial nucleic acid comprises at least one coding region encoding at least one polypeptide comprising or consisting of at least one Norovirus capsid protein, and/ or a fragment or variant thereof.
  • the artificial nucleic acid molecule according to the invention comprises at least one coding region encoding the Norovirus capsid protein VPI or VP2.
  • the artificial nucleic acid encodes Norovirus capsid protein VPI.
  • a reference to a Norovirus capsid protein VPI equals a reference to a VPI capsid protein derived from a Norovirus, a Norovirus VPI, capsid protein, capsid protein VPI, major capsid protein, major capsid protein VPI, major capsid region, major viral capsid protein, or VPI capsid protein.
  • the invention relates to an artificial nucleic acid comprising or consisting of at least one coding region encoding at least one polypeptide selected from the group consisting of Norovirus capsid protein VPI (also termed “Major capsid protein” or “capsidprotein”), Norovirus capsid protein VP2 (also termed “Minor capsid protein”) and/ or a Norovirus non-structural protein, such as NSI/NS2 (also termed p48 or Nterm (amino terminal protein)), NS3 (also termed NTPase or Nucleoside triphosphatase).
  • VPI also termed “Major capsid protein” or “capsidprotein”
  • Norovirus capsid protein VP2 also termed “Minor capsid protein”
  • NSI/NS2 also termed p48 or Nterm (amino terminal protein)
  • NS3 also termed NTPase or Nucleoside triphosphatase
  • NS4 also termed p22 or 3A-like protein
  • NS5 also termed VPg or Genome-linked viral protein
  • NSB also termed Pro or Proteinase
  • NS7 also termed Pol or NA-dependent NA polymerase
  • vaccines and/or compositions contain VPI proteins and/or VP2 proteins.
  • each vaccine and/or composition contains VPI and/or VP2 proteins from only one Norovirus genogroup giving rise to a monovalent vaccine.
  • the term "monovalent" means the antigenic proteins are derived from a single Norovirus genogroup.
  • the vaccines and/ or compositions contain VPI and/ or VP2 from a virus strain of genogroup I (e.g. VPI and VP2 from Norwalk virus).
  • the vaccines and/ or compositions are comprised of predominantly VPI proteins.
  • the antigen is a mixture of monovalent vaccines and/or compositions wherein the composition includes vaccines and/or compositions comprised of VPI and/or VP2 from a single Norovirus genogroup mixed with vaccines and/or compositions comprised of VPI and/or VP2 from a different Norovirus genogroup taken from multiple viral strains (e.g. a Norovirus from Genogroup IV.2 and a Norovirus from Genogroup I.I).
  • the composition can contain monovalent vaccines and/ or compositions from one or more strains of Norovirus genogroup I together with monovalent vaccines and/ or compositions from one or more strains of Norovirus genagroup II.
  • the Norovirus vaccines and/ or composition mixture is composed of the Norovirus from Genogroup IV.2 and Norovirus from Genogroup 1.1 or from different genus or species of a Norovirus from Genogroup IV.2.
  • the vaccines and/ or compositions are comprised of predominantly VPI proteins.
  • the antigen is a mixture of monovalent vaccines and/or compositions wherein the composition includes vaccines and/or compositions comprised of VPI and/or VP2 from a single Norovirus genogroup mixed with vaccines and/or compositions comprised of VPI and/or VP2 from a different Norovirus genogroup taken from multiple viral strains (e.g.
  • the composition can contain monovalent vaccines and/ or compositions from one or more strains of Norovirus genogroup I together with monovalent vaccines and/or compositions from one or more strains of Norovirus genogroup II.
  • the Norovirus vaccines and/or composition mixture is composed of the Norovirus from Genogroup 11.4 and Norovirus from Genogroup I.I or from different genus or species of a Norovirus from Genogroup 11.4.
  • the amino acid seguence of the at least one antigenic peptide or protein may be selected from any peptide or protein derived from a capsid protein VPI, capsid protein VP2, NSI/NS2, NS3, NS4, NS5, NSB, or NS7 of a Norovirus or a fragment or variant thereof.
  • the vaccines and/or compositions may be multivalent vaccines and/or compositions that comprise, for example, VPI and/or VP2 proteins from one Norovirus genogroup intermixed with VPI and/or VP2 proteins from a second Norovirus genogroup, wherein the different VPI and VP2 proteins are not chimeric VPI and VP2 proteins, but associate together within the same capsid structure to form immunogenic Vaccines and/or compositions.
  • Multivalent vaccines and/ or compositions may contain vaccines and/ or composition antigens taken from two or more viral strains.
  • the composition can contain multivalent vaccines and/or compositions comprised of capsid monomers or 5 multimers from one or more strains of Norovirus genogroup I together with capsid monomers or multimers from one or more strains of Norovirus genogroup II.
  • the Norovirus vaccines and/ or composition mixture is composed of the Norovirus from Genogroup IV.2 and Narovirus from Genogroup I.I or from different genus or species of a Narovirus from Genogroup IV.2.
  • the vaccines and/or compositions may be multivalent vaccines and/or ID compositions that comprise, for example, VPI and/or VP2 proteins from one Norovirus genogroup intermixed with VPI and/or VP2 proteins from a second Norovirus genogroup, wherein the different VPI and VP2 proteins are not chimeric VPI and VP2 proteins, but associate together within the same capsid structure to form immunogenic Vaccines and/or compositions.
  • the term "multivalent” means that the antigenic proteins are derived from two or more Norovirus genogroups.
  • Multivalent vaccines and/or compositions may contain vaccines and/or composition antigens taken from two or more viral strains.
  • the composition can contain multivalent vaccines and/or compositions comprised of capsid monomers or multimers from one or more strains of Norovirus genogroup I together with capsid monomers or multimers from one or more strains of Norovirus genogroup II.
  • the Norovirus vaccines and/or composition mixture is composed of the Norovirus from Genogroup 11.4 and Norovirus from Genogroup I.I or from different genus or species of a Narovirus from Genogroup 11.4.
  • the artificial nucleic acid may comprise 2, 3, 4, 5, G, 7, 8, 9, ID or more coding regions encoding at least one polypeptide selected from the group consisting of Norovirus capsid protein VPI, Norovirus capsid protein VP2, NS1/NS2. NS3, NS4, NS5, NSB, or NS7, and/ or a fragment or variaot of any of these proteins.
  • compositions or vaccines of the present invention comprise a multivalent vaccine, e.g.,5 comprising a polynucleotide which encodes at least two different VPI, for example a Norovirus capsid protein VPI from a GII.4 strain and a Norovirus capsid protein VPI from a Gl.l strain.
  • a multivalent vaccine e.g.,5 comprising a polynucleotide which encodes at least two different VPI, for example a Norovirus capsid protein VPI from a GII.4 strain and a Norovirus capsid protein VPI from a Gl.l strain.
  • compositions or vaccines comprises a multivalent vaccine, e.g., comprising two different polynucleotides, whereby one polynucleotide encodes for example a Norovirus capsid protein VPI from a GII.4 strain and the other polynucleotide encodes a Norovirus capsid protein VPI from a Gl.l strain.
  • the compositions or vaccines of the present invention may comprise a multivalent vaccine, e.g., comprising a polynucleotide which encodes at least two different VPI, for example a Norovirus capsid protein VPI from two different GII.4 strains.
  • compositions or vaccines comprise a multivalent vaccine, e.g., comprising two different polynucleotides, whereby one polynucleotide encodes for example a Norovirus capsid protein VPI from two different GII.4 strains.
  • the composition or the vaccine is multivalent, i.e. compositions or vaccines of the present invention may vary in their valency. Valency refers to the number of antigenic components in the composition or vaccine.
  • the compositions or vaccines are monovalent.
  • the compositions or vaccines are divalent or bivalent.
  • the compositions or vaccines are trivalent.
  • the compositions or vaccines are teravalent.
  • the compositions or vaccines are multi-valent.
  • Multivalent vaccines may comprise at least 2, 3, 4, 5, G, 7, 8, 9, 10, II, 12, 13, 14, 15, IB, 17, 18, 19, 20, 21, 22, 23, 24, 25, 2B, 27, 28, 29, 3D, 31, 32, 33, 34, 35, 3B, 37. 38, 39, 40, 41, 2, 43, 44, 45. 4B. 47, 48, 49, 50. 51, 52, 53, 54, 55, 5B, 57, 58, 59, BO. Bl, B2, B3, B4, B5, BB, B7, B8, B9, 70, 71, 72, 73.74, 75, 7B. 77, 78. 79, 80, 81. 82, 83, 84, 85. 8B.
  • compositions or vaccines may be on a single polynucleotide or on separate polynucleotides.
  • multivalent vaccines may comprise or at least 10, 15, 20, 30. 40 or 50 or 100 or more antigens or antigenic moieties (e.g., antigenic peptides, etc.).
  • multivalent vaccines may comprise 2-10, 10-15, 15-20, 20-50, 50-100 or 100-200 or more antigens or antigenic moieties (e.g., antigenic peptides, etc.).
  • the multivalent composition or vaccine comprises about 30 to about 5D antigens or antigenic moieties.
  • the open reading frame of the one or more NA polynucleotides encodes at least 2, 3, , 5, G, 7, 8, 9 or 10 or at least 10, 15, 20 or 50 or 2-10, 10-15, 15-20, 20-50, 50-100 or 100-200 antigenic polypeptides.
  • the composition comprises at least two mRNA sequences, wherein at least one mRNA sequence encodes at least one antigenic peptide or protein, i.e. VPI, is derived from a GII.4 Norovirus and at least one mRNA sequence encodes at least one antigenic peptide or protein, derived from Norovirus VPI, is derived from another GII.4 Norovirus.
  • each mRNA sequence encodes at least one different antigenic peptide or protein derived from proteins of different Noroviruses.
  • each mRNA sequence encodes at least one antigenic peptide or protein, derived from Norovirus VPI, of different GII.4 Noroviruses or different Gl.l Noroviruses, or a combination thereof.
  • the invention relates to a composition comprising or consisting of 2, 3, 4, 5, G, 7, 8, 9, 10, II, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25.2B.27, 28, 29, 30, 31.
  • the open reading frame of the one or more RNA polynucleotides encode at least I0, 15, 2D, 3D, 40 or 5D or IDD antigenic polypeptides.
  • the open reading frame nf the one or more RNA polynucleotides encode 2- ID, ID-I5. 15-2D, 20-5D, 5D-IDD or I00-2D0 antigenic polypeptides.
  • the present invention is based on the surprising finding that the at least one Norovirus protein comprised in the at least one polypeptide encoded by the artificial nucleic acid as described herein can efficiently be expressed in a mammalian cell. It was further unexpectedly found that the artificial nucleic acid is suitable for eliciting an immune response against Norovirus in a subject.
  • the present invention is based on the surprising finding that mRNA-based or artificial nucleic acid vaccines comprising mRNA nr artificial nucleic acid sequences encoding different antigens of a Norovirus (particularly Norovirus capsid protein VPI) were extremely effective in inducing an antigen-specific immune response against Norovirus. Furthermore, the inventors surprisingly found that many mRNA sequences encoding different antigens of different Noroviruses can be effectively combined in ooe mRNA-based vaccine.
  • the artificial nucleic acid of the invention comprises at least one coding region encoding at least one polypeptide derived from a Norovirus and/or a fragment or variant thereof.
  • the artificial nucleic acid of the invention comprises at least one coding region encoding at least one0 polypeptide selected from the group consisting of a non-structural protein derived from a Norovirus and/or a capsid protein derived from a Norovirus, and/ or a fragment or variant thereof.
  • the artificial nucleic acid of the invention comprises at least one coding region encoding at least one polypeptide selected from the group consisting of Norovirus non-structural proteins NSI/NS2, NS3, NS4, NS5, NSB, NS7,5 Norovirus capsid protein VPI and Norovirus capsid protein VP2, and/ or a fragment or variant thereof.
  • the term “Norovirus” comprises any Norovirus, irrespective of strain or origio.
  • the term “Norovirus” comprises a Norovirus strain selected from the group consisting of Genogroup I, Genogroup II, Genogroup0 III, Genogroup IV, or Genogroup V (abbreviated as Gl, Gil, Gill, GIV or GV. respectively).
  • Neovirus comprises a Norovirus strain selected from the group consisting of
  • Genogroup I genotype I (abbreviated as Gl.l), GI.2, GI.3, GI.4, GI.5, GI.G, GI.7, GI.8, GI.9. GI.IO, GUI, GI.I2, GI.I3. GI.I4, GI.I5.
  • Genogroup II genotype I (abbreviated as Gll.l), GII.2, GII.3. GII.4, GII.5, GII.G, GII.7, GII.8, GII.9. GII.IO, Gll.ll. GII.I2. GII.I3,
  • Genogroup II! genotype I (abbreviated as Glll.l), GIII.2, GIII.3, and/or GIII.4;
  • Genogroup IV genotype I (abbreviated as GIV.I). GIV.2, GIV.3, and/or GIV.4;
  • Genogroup V genotype I (abbreviated as GV.I), GV.2, GV.3. and/or GV.4;
  • the nucleic acid of the invention preferably a VPI nucleic acid derived from a Norovirus, is derived from a Norovirus selected from the group consisting of
  • Genogroup I genotype I (abbreviated as Gl.l), GI.2, GI.3, GI.4, GI.5, GI.G, GI.7. GI.B. GI.B, GI.IO, GUI. GI.I2. GI.I3. GI.I4, GI.I5, ID GLIB and/or GI.I7;
  • Genogroup II genotype I (abbreviated as Gll.l), GII.2, GII.3, GII.4, GII.5, GII.G, GII.7, GII.8, GII.9, GII.ID, Gll.ll. GII.I2, GII.I3.
  • Genogroup III genotype I (abbreviated as Glll.l), GIII.2, GIII.3, and/or GIII.4;
  • Genogroup IV genotype I (abbreviated as GIV.I), GIV.2, GIV.3, and/or GIV.4; and
  • Genogroup V genotype I (abbreviated as GV.I), GV.2, GV.3, and/ or GV.4.
  • VPI nucleic acids can be derived from 2, 3, , 5, B, 7, 8, B, or IQ, or more of any of the above Noroviruses from different Genogroups and/or different Genotypes.
  • a reference to Gl.l to GI.I7 means a reference to Gl.l, GI.2, GI.3, GI.4. GI.5, GI.B, GI.7, GI.8, GI.B, GI.IO, GUI, GI.I2, GI.I3, GI.I4, GI.I5, GLIB and/or GI.I7; a reference to Gll.l to GII.24 means a reference to Gll.l, GII.2, GII.3, GII.4, GII.5, GII.G. GII.7, GII.8.
  • a reference to Glll.l to GIII.4 means to a reference to Glll.l, GIII.2, GIII.3, and/or GIII.4; a reference to GIV.I to GIV.4 means to a5 reference to GIV.I, GIV.2, GIV.3, and/or GIV.4; a reference to GV.I to GV.4 means to a reference to GV.I, GV.2, GV.3, and/or GV.4.
  • the term "Norovirus” comprises a Norovirus strain selected from the group consisting of a GII.4 Norovirus and/or a Gl.l Norovirus.
  • the term “Norovirus” as used herein refers to Norovirus GII.4 CIN-I (also termed Norovirus Hu/GII.4/D3IB33/USA/2DD3 or having Accession No. JD3B58ID.I), GII.4 (Accession No; AY5D2D23.I), GII.4 CIN-0 D02 and/ or GII.4 Sydney.
  • the Norovirus is a GII.4 Sydney Norovirus or a GII.4 Sydney 2DI2 Norovirus.
  • the term “Norovirus” comprises a Norovirus strain selected from the group consisting of a GII.4 Norovirus and/or a Gl.l Norovirus.
  • the term “Norovirus” as used herein refers to Norovirus GII.4-03IG93-USA- 2D03, Norovirus GII.4/Farmington Hills/20D2/DSA, Norovirus Gll.4-2006b 092895-0-2008, Norovirus GII.4-GZ20I0-L87-5 Guangzhou-2011, Norovirus GII.4-USA-I997, Norovirus GU-OSA-IGBB-Capsidprotein.
  • Neovirus comprises a Norovirus strain selected from the group consisting of Norovirus Hu/GII.4/Dijon/E872/2DD2/F A. Norovirus Hu/GII.4/MDI2D-l2/ie87/USA, Norovirus Hu/GIIJ/7EK/Hawaii/l97l/USA, Norovirus Hu/GII.B/CHDC4073/I984/USA, Norovirus Hu/GII.4/Hiroshima/IB/2G0l/JPN, Norovirus Hu/GII.4/Hiroshima/67/2DDG/JPN.
  • Hu/Gll.4/Wellington/I995/OSA Norovirus Hu/GII.4/Henry/2000/OSA, Norovirus Hu/GII.4/SSCS/2005/USA, Norovirus GII/Hu/IN/200B/GII.P4_GII.4_ Yerseke200Ba/Pune-PC2l, Norovirus Hu/GI.I/P7-587/2007/Stromstad/Swed.n, Norovirus Hu/GI.2/Leuven/2003/BEL Nurovirus Hu/GII.7/NSW743L/2008/AUS.
  • Hu/GGII.4/ iddelburg007/20D4/NL Norovirus Hu/GII-4/Matsudo/02l07l/2D02/JP, Norovirus Hu/GII- 4/ aiso/03055B/2003/JP.
  • Norovirus Hu/GII-4/Awa/040354/2004/JP Norovirus Hu/GII.4/Apeldoorn317/2007/NL
  • Norovirus Hu/GII.4/RotterdamP200/2005/NL Norovirus Hu/Gll.4/Stockholm/I98B5/2008/SE.
  • Norovirus Hu/GII.B/DC040B2VLP/2004/JP Norovirus Hu/GII.4/HSI94/2DD9/US.
  • Norovirus Hu/GII.I2/HS2IO/20IO/OSA Norovirus Hu/GI.I/8Flla/l9G8/0SA.
  • Norovirus Hu/Gll.4/CHDC5I9I/I974/USA Norovirus Hu/GII.4/N7G/20!0/HuZhou.
  • Norovirus Hu/GII.B/S9c/I97G/SEN Norovirus Hu/GII L45/I978/MYS.
  • Norovirus Hu/GII.4/NIHIC9/20ll/DSA Norovirus Hu/Gll.4/Cll0/I378/GUF, Norovirus Hu/GII.4/HSBB/200l/USA.
  • Norovirus Hu/GIU2/Shelby/2D09/DSA Norovirus Hu/GI.7/TCH-OGD/USA/2003, Norovirus Hu/GII.I/Ascension2D8/2DI0/USA, Norovirus Hu/GII.I3/VAI73/2DI0/USA, Norovirus Hu/GII.2l/Salisburyl50/20ll/DSA.
  • Norovirus Hu/Gll.4/I997/USA Norovirus Hu/GII.4/Farmington Hills/2BD4/USA, Norovirus Hu/GII.4/MinervB/2D0B/USA,
  • Norovirus Hu/GII.4/PR328/2DI3/ITA Norovirus Hu/GII.PI7_GII.I7/PRBB8/2DI5/ITA, Norovirus Hu/GII.4/Alb E rtaSPI/2DI3/CA, Norovirus HU/GII.4/CDDDD7BB2 2DII/UK.
  • Norovirus Hu/GII.4/MBlbournBBB23/2DIB/AUS Norovirus GII/Hu/JP/2DIB/GII.PIB_GII.4_ SydnBy2DI2/DHIB002, Norovirus Hu/GII/JP/20IB/GII.PIB_GII.4_ SydnBy2DI2/ awasakil94.
  • the artificial nucleic acid is derived from a Norovirus selected from the group consisting of genogroup I Norovirus, genogroup II Norovirus, genogroup III Norovirus, gBnogroup IV Norovirus, and genogroup V Norovirus; preferably the artificial nucleic acid is derived frrnn a Norovirus selscted from the group consisting of a Gl.l to GI.I7 Norovirus, Gll.l to GII.24 5 Norovirus, Glil.l to GIII.4 Norovirus. GIV.I to GIV.4 Norovirus and GV.I to GV.4 Norovirus; more prefErably.
  • a Norovirus selected from the group consisting of genogroup I Norovirus, genogroup II Norovirus, genogroup III Norovirus, gBnogroup IV Norovirus, and genogroup V Norovirus
  • the artificial nucleic acid is derived frrnn
  • the artificial nucleic acid is derived from a Norovirus selected from the group consisting of Gl.l Norovirus and GII.4 Norovirus, even ⁇ preferably, the artificial nucleic acid is derived from a GII.4 Norovirus, still more preferably, the artificial nucleic acid is derived from a GII.4 CIN-I Norovirus or a GII.4 Sydney Norovirus or a GII.4 Sydney 2DI2 Norovirus.
  • the artificial nucleic acid is derived from a Norovirus selected from the group consisting of Norovirus GII.4-03IB93-USA-2D03, Norovirus GIU/Farmington Hills/2DD2/DSA, Norovirus GII.4-2DDBb 0928B5-DSA-2008, Norovirus GII.4-GZ2DI0-L87-Guangzhou-2DII. Norovirus GII.4-USA-I997, Norovirus GI.I-DSA-l9G8-Capsidprotein.
  • a Norovirus selected from the group consisting of Norovirus GII.4-03IB93-USA-2D03, Norovirus GIU/Farmington Hills/2DD2/DSA, Norovirus GII.4-2DDBb 0928B5-DSA-2008, Norovirus GII.4-GZ2DI0-L87-Guangzhou-2DII. Norovirus GII.4-USA-I997, Noro
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises at least one Norovirus protein.
  • the RNA genome of Norovirus typically encodes a plurality of structural and non-structural proteins. Translation of Norovirus RNA typically leads to a precursor protein comprising a plurality of individual viral (structural and nonstructural) proteins (or precursor of these proteins) in one polypeptide chain, which is typically referred to as "polyprotein” or "precursor protein".
  • a Norovirus polyprotein typically comprises amino acid sequences that are target sites for enzymes that specifically cleave the polyprotein in order to yield fragments of the polyprotein, wherein the fragments preferably comprise an individual Norovirus protein or two or more Norovirus proteins, or a fragment or variant thereof.
  • the term "polyprotein” may also refer to a polypeptide chain comprising the amino acid sequences of at least two individual Norovirus proteins, or a fragment or variant thereof. Cleavage of a Norovirus polyprotein preferably occurs between individual Norovirus proteins (e.g. between the capsid protein VPI and the capsid protein VP2, or fragments or variants thereof.
  • an individual Norovirus protein, or a fragment or variant thereof, e.g. as obtained from a polyprotein by cleavage is preferably referred to as "mature Norovirus protein".
  • the term "mature Norovirus protein” is not limited to an individual Norovirus protein, or a fragment or variant thereof, which was generated by cleavage of a polyprotein, but also comprises an individual Norovirus protein of another origin, such as an individual Norovirus protein expressed recombinantly from an artificial nucleic acid.
  • a mature Norovirus protein lacks an amino acid sequence that is typically present in a corresponding amino acid sequence encoding said Norovirus protein in a Norovirus polyprotein (precursor protein) and wherein said amino acid sequence lacking in the mature Norovirus protein preferably corresponds to an amino acid sequence, which is usually removed by cleavage during processing of a Norovirus polyprotein.
  • an amino acid sequence, which is a target site for a protease may be present in a Norovirus polyprotein, but may be absent from a mature Norovirus protein derived from said Norovirus polyprotein.
  • the artificial nucleic acid comprising at least one coding region encoding at least one polypeptide comprises at least one Norovirus capsid protein VPI or Norovirus capsid protein VP2 and/or a fragment or a variant thereof. In another embodiment, the artificial nucleic acid comprising at least one coding region encoding at least one polypeptide comprises at least one Norovirus capsid protein VPI and/ or a fragment or variant thereof. In a further embodiment, the artificial nucleic acid comprises a Norovirus capsid protein VPI and/or a fragment or variant thereof.
  • Nevirus protein may refer to any amino acid encoded by a Norovirus nucleic acid.
  • a Norovirus capsid protein VPI or VPI protein is preferred.
  • a “Norovirus protein” may be any polypeptide comprising or consisting of an amino acid sequence according to any one of the following amino acid sequences from Genbank, or a fragment or variant of any of these sequences as provided in Table I (Column 2: “NC8I or Genbank Accession No.") and Table 3 (Column 2; "NCBI or Genbank Accession No.”).
  • the term “Norovirus protein” as used herein comprises an individual structural or non-structural Norovirus protein.
  • a Norovirus protein in the meaning of the present invention may be a protein selected from the group consisting of Norovirus capsid protein VPI. Norovirus capsid protein VP2. and a Norovirus non-structural protein (NS). such as NSI/NS2, NS3, NS4, NS5, NSB, or NS7.
  • NS Norovirus non-structural protein
  • Nemovirus protein as used herein is a Norovirus capsid protein VPI.
  • Nemovirus protein as used herein is a Norovirus capsid protein derived from a GII.4 Norovirus.
  • Nonovirus protein may also refer to an amino acid sequence corresponding to an individual Norovirus protein as present in a Norovirus polyprotein (precursor protein). Said amino acid sequence in the polyprotein may differ from the amino acid sequence of the corresponding amino acid sequence of the respective mature Norovirus protein (i.e. after cleavage/processing the polyprotein).
  • the corresponding amino acid sequence comprised in the polyprotein may comprise amino acid residues that are removed during cleavage/processing of the polyprotein (such as a signal sequence or a target site for a protease) and that are no longer present in the respective mature Norovirus protein.
  • Neovirus protein comprises both, the precursor amino acid sequence comprised in a Norovirus polyprotein (i.e. as part of a polypeptide chain optionally further comprising other viral proteins) as well as the respective mature individual Norovirus protein.
  • Non-virus capsid protein VPI may refer to an amino acid sequence in a Norovirus polyprotein corresponding to the precursor sequence of Norovirus capsid protein VPI (comprising, for example, a (C-terminal) signal sequence) as present in a Norovirus polyprotein as well as to a mature (separate) Norovirus capsid protein VPI (no longer comprising, for example, a (C-terminal) signal sequence).
  • any numbering used herein - unless stated otherwise - relates to the position of the respective amino acid residue in a Norovirus polyprotein (precursor protein), wherein position "I" corresponds to the first amino acid residue, i.e. the amino acid residue at the N-terminus of a Norovirus polyprotein.
  • the numbering with regard to amino acid residues refers to the respective position of an amino acid residue in a Norovirus polyprotein, which is preferably derived from a Norovirus strain selected from the group consisting of Genogroup II Dr Genogroup I (abbreviated as Gil, or Gl, respectively), more preferably from the group consisting of Genogroup II genotype 4 (abbreviated as GII.4) or Genogroup I genotype I (abbreviated as GII.4 and Gl.l), or even more preferably the strain is selected from strain Norovirus strain GII.4 CIN-I or CIN-DD2.
  • a Norovirus polyprotein which is preferably derived from a Norovirus strain selected from the group consisting of Genogroup II Dr Genogroup I (abbreviated as Gil, or Gl, respectively), more preferably from the group consisting of Genogroup II genotype 4 (abbreviated as GII.4) or Genogroup I genotype I (abbreviated as GII.4 and Gl.l), or even more
  • Preferred Norovirus protein in the context of the invention may be any polypeptide comprising or consisting of an amino acid sequence according to any one of the following amino acid sequences from Genbank, or a fragment or variant of any of these sequences: AC0550B8, AFS33552, AFS33555, AFX7IGB5, BAI49904, BAI4B9I4, BAS02D83, CRL4G958, CRL4B973, ADB27D27, AGI9B397, AGXDI095, AGX0I098, A I300B0, AGL984I3, ACT7BI45, ACT7BI48, ACT7BI5I, AED02034, AEXI0549, AFJ2I448, AFN0B72B, AFN0B727, AFN0B73I, AFNDB732, AFN0B733, AFN0B735, AFV0877I, AFV08777, AFV08795, AFX95940, AGI99552, AGK
  • BAF745D8 BAF74509, BAF745I2, BAF745I7, BAF745ZI, BAF95499, BAF955DI.
  • PrefErrEd examples of Noraviruses which may be used for providing the nucleic acid molecules of the invention may include IB Norovirus GII.4-03IB93-USA-ZBB3.
  • Norovirus GI/Hu/JP/Z0D7/GI.P8_GI.8/Nagoya/KY53l Norovirus Hu/GII.4/SJTDHI/CHN/ZDI4, Norovirus Hu/GII.4/variant Sydney Z0I2/FRA, Norovirus Hu/GII-4/Hokkaido4/ZDDB/JP.
  • Norovirus Hu/ll.4/220DBGI/HK/2DI0 Norovirus Hu/ll.4/220DBGI/HK/2DI0.
  • Norovirus pig/GII.II/FI8-IO/2D05/CAN Norovirus5 Hu/GII.4/Wellington/l995/USA, Norovirus Hu/GI /h y/ZOOO/DSA, Norovirus Hu/GII.4/SSCS/2DD5/USA.
  • Norovirus I2-X-2/20I2/GII.P22/GII.5 Norovirus Hu/GII.4/ obBQ34/2DDG/JP.
  • Hu/GGII.4/MiddelburgDD7/2Q04/NL Norovirus Hu/GII-4/Matsudo/Q2l07l/2Q02/JP
  • Norovirus Hu/GII- 4/Kaiso/D3D55G/2QD3/JP Norovirus Hu/GII-4/Awa/04D354/2G04/JP
  • Norovirus Hu/GII.4/Apeldoorn3l7/2Q07/NL Norovirus Hu/GII.2/Rotterdam39E/2002/NL
  • Hu/BII.4/Kenepuru/NZ327/2DDB/NZL Norovirus Hu/GI /Rathmines/NSW287R/2007/A0S, Norovirus Hu/GII.4/Turramurra/NSW892U/20D9/AUS, Norovirus HU/GII.4/SEOUI/0389/2D09/KDR, Norovirus Hu/GII.4/Seoul/0945/2D09/ DR, Norovirus Hu/GII.I2/Shelby/2009/LISA.
  • Norovirus GII.I2 Snow Mountain virus. Human calicivirus strain Mslksham.
  • Norovirus genogroups such as a genogroup I.I (Norwalk virus) and 11.4 (f.e. Houston virus) or other commonly circulating strains, or synthetic constructs representing combinations or portions thereof are preferred in some embodiments.
  • New strains of Noroviruses are routinely identified (Centers for Disease Control, Morbidity and Mortality Weekly Report, 56(33):842-846 (2007)) and consensus sequences of two or more viral strains may also be used to express Norovirus antigens.
  • the at least one polypeptide encoded by the at least one coding region of the artificial nucleic acid may consist of an individual Norovirus protein, the amino acid sequence of which does typically not comprise an N- terminal methionine residue. It is thus understood that the phrase "polypeptide consisting of Norovirus protein " relates to a polypeptide comprising the amino acid sequence of said Norovirus protein and - if the amino acid sequence of the respective Norovirus protein does not comprise such an N-terminal methionine residue - an N-terminal methionine residue.
  • the inventive artificial nucleic acid comprises at least one coding region encoding at least one polypeptide comprising or consisting of at least one Norovirus protein as described herein, wherein the at least one Norovirus protein comprises an amino acid sequence according to any one of SEQ 10 NOs: 1-4410, or a fragment or variant of any of these sequences.
  • sequence refers to any one of SEQ ID Nos: I-39B90, 337I3-3B746.
  • the artificial nucleic acid of the invention comprises at least one encoded polypeptide comprising
  • amino acid sequences having, in increasing order of preference, at least 50%, 51%, 52%, 53%, 54%, 55%, 50%, 57%, 58%, 58%, 80%, 81%, 82%, 83%, 84%, 85%, 66%, 87%, 88%, 83%, 70%, 71%, 72%, 73%,
  • the artificial nucleic acid of the invention comprises at least one coding region comprising
  • nucleic acid sequences having, in increasing order of preference, at least 50%. 51%, 52%, 53%, 54%, 55%, 56%. 57%, 58%, 50%, 60%. 61%, 62%, 63%.
  • nucleic acid sequences which are capable of hybridizing with a nucleic acid sequence comprising a sequence as shown in SEQ ID NOs: 4411-39690, 39713-39746, and/or to a nucleic acid encoding a polypeptide having a sequence as shown in SEQ ID NOs: 1-4410, and/or
  • a fragment of a protein or a variant thereof encoded by the at least one coding sequence of the artificial nucleic acid according to the invention may typically comprise an amino acid sequence having a sequence identity of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%, preferably of at least 70%, more preferably of at least 80%, even more preferably at least 85%, even more preferably of at least 90% and most preferably of at least 95% or even 97%, with an amino acid sequence of the respective naturally occurring full-length protein or a variant thereof, preferably as disclosed in Table I, column I. column 2 or column 3, more preferably as disclosed in Table 3, column I, column 2, column 3.
  • the at least one coding sequence of the artificial nucleic acid sequence according to the invention preferably encodes Norovirus proteins selected from the proteins provided in Table I, or a fragment or variant thereof. Any Norovirus protein provided in Table I, or any a fragment or variant thereof, can cause an immune response when administered to an individual. Therefore, all Norovirus proteins provided in Table I and Table 3 can be considered as preferred Norovirus antigens in the context of the present invention.
  • the at least one coding sequence of the artificial nucleic acid sequence of the present invention encodes a Norovirus protein or peptide, or a fragment or variant thereof, wherein the Norovirus protein or peptide is an antigen selected from the antigens listed in Table I.
  • each row corresponds to a Norovirus protein or antigen as identified by the respective gene name (first column, column I "Name”) and the database accession number of the corresponding protein (second column, column 2 "NCBI or Genbank Accession No.”).
  • the third column, column 3 ("A") in Table I indicates the SED ID NOs corresponding to the respective amino acid sequence as provided herein.
  • the SEQ ID NOs corresponding to the nucleic acid sequence of the wild type nucleic acid sequence encoding the Norovirus protein or peptide is indicated in the fourth column, column 4 ("B"),
  • the fifth column, column 5 (“C") provides the SEQ ID NOs corresponding to modified nucleic acid sequences of the nucleic acid sequences as described herein that encode the Norovirus protein or peptide preferably having the amino acid sequence as defined by the SED ID NDs indicated in the third column ("A”) or by the database entry indicated in the second column ("NCBI or Denbank Accession No.”).
  • the at least one coding sequence of the artificial nucleic acid sequence of the present 5 invention encodes at least one Nnrovirus protein or peptide which is derived from Norovirus polyprotein, or a fragment or variant thereof, wherein the Norovirus polyprotein is selected from the Nnrovirus polyprotein amino acid sequences listed in Table I.
  • each row corresponds to a Norovirus polyprotein as identified by the respective gene name (first column "Name”, derived from NCBI or Genbank) and the database accession number of the corresponding protein (second column "NCBI or Benbank Accession No.”).
  • the third column (“A") in Table I indicates the SED ID NDs corresponding to the respective amino acid ID sequence as provided herein.
  • the SED ID NDs corresponding to the nucleic acid sequence of the wild type nucleic acid encoding the Norovirus protein or peptide is indicated in the fourth column ("B").
  • the fifth column (“C") provides the SED ID NDs corresponding to modified nucleic acid sequences of the nucleic acids as described herein that encode the Norovirus protein or peptide preferably having the amino acid sequence as defined by the SEQ ID NDs indicated in the third column (“A”) or by the database entry indicated in the second column ("NCBI or Genbank Accessioo No.”).
  • Norovirus polyprotein and nucleic acid sequences according to SEQ ID NDs: 397I3- 3374G.
  • the Norovirus protein or peptide is derived from a Norovirus capsid protein VPI according to SEQ IDD NOs: 1-4410.
  • the at least one coding sequence of the artificial nucleic acid sequence of the present invention encodes at least one Norovirus protein or peptide which is derived from Norovirus capsid protein VPI, Dr a fragment or variant thereof, wherein the Norovirus capsid protein VPI is selected from the Norovirus capsid protein VPI amino acid5 sequences listed in Table I.
  • each row corresponds to a Norovirus capsid protein VPI as identified by the respective gene name (first column "Name”) and the database accessinn number of the corresponding protein (second column "NCBI or Genbank Accession No").
  • the third column (“A") in Table I indicates the SEQ ID NDs corresponding to the respective amino acid sequence as provided herein.
  • the SED ID NDs corresponding to the nucleic acid sequence of the wild type RNA encoding the Norovirus antigen is indicated in the fourth column ("B").
  • the fifth column (“C") provides the SED ID NDs corresponding to modified nucleicD acid sequences of the RNAs as described herein that encode the Norovirus antigen preferably having the amino acid sequence as defined by the SEQ ID NDs indicated in the third column ("A") or by the database entry indicated in the second column ("NCBI or Genbank Accession No").
  • the inventive artificial nucleic acid comprises or consists of at least one coding sequence5 encoding at least one Norovirus protein or peptide as described herein.
  • the inventive artificial nucleic acid comprises or consists of a coding sequence according to any one of SEQ ID NOs: 44II-3BG9D. 39713-30746, or a homolag, fragment or variant of any of these sequences (see Table I, column “B” and “C”).
  • a nucleic acid molecule of the invention is at least 15, 2D, 25 or 30 nucleotides in length. 5 Preferably, it hybridizes under stringent conditions to a nucleic acid molecule comprising a nucleotide sequence of the nucleic acid molecule of the present invention or used in the methods of the present invention.
  • the nucleic acid molecule is preferably at least 20, 30, 50, IDD, 250 or more nucleotides in length.
  • the present invention thus provides artificial nucleic acid sequences comprising at least one coding ID sequence, wherein the coding sequence encoding Norovirus capsid protein VPI comprises or consists any one of the nucleic acid sequences defined in Table I, preferably in the fourth or fifth column (column "B” or “C”, respectively) of Table I, or a fragment or variant of any one of these sequences.
  • the nucleic acid sequence comprises or consists of at least one coding sequence encoding 15 Norovirus capsid protein VPI according to SEQ ID NDs: 4411-39690, 39713-39746.
  • the nucleic acid sequence according to the invention comprises at least one coding sequence encoding Norovirus capsid protein VPI comprising a nucleic acid sequence selected from sequences being identical or at least 50%. 60%, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, orD 99% identical to the nucleic acid sequences as disclosed in Table I, preferably in the fourth or fifth column (column "B" or "C", respectively) of Table I or a fragment or variant thereof.
  • the present invention provides an nucleic acid sequence as defined herein comprising at least one coding sequence encoding at least one Norovirus peptide or protein derived from Norovirus capsid5 protein VPI, wherein the coding sequence comprises or consists of any one of the (modified) nucleic acid sequences defined in the Column "C" of Table I, or of a fragment or variant of any one of these sequences.
  • the inventive artificial nucleic acid comprises or consists of at least one coding sequence encoding at least one Norovirus protein or peptide as described herein, wherein the at least one Norovirus protein comprises0 an amino acid sequence according to any one of SEQ ID NDs: 1-4410, or a homolog, fragment or variant of any of these sequences (see Table I, third column, column "A").
  • the inventive artificial nucleic acid comprises at least one coding sequence encoding at least one protein or peptide derived from a Norovirus, or a fragment Dr variant thereof, wherein the Norovirus protein or peptide preferably comprises or consists of any one of the amino acid sequences defined in the third column (column "A") of Table I, or a fragment or variant of any one of these sequences.
  • the at least one5 coding sequence preferably encodes a Norovirus protein comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-4410.
  • the at least one coding sequence of the nucleic acid sequence according to the invention comprises or consists of an nucleic acid sequence identical to or having a sequence identity of at least 5%, 10%, 20%, 30%, 40%, 50%, G0%, 70%, 80%, 85%, 86%, 87%, 88%, 83%, 80%, 91%. 02%. 93%. 94%, 95%.
  • 9G%, 97%, 98%, or 99% preferably of at least 70%, more preferably of at least 80%, even more preferably at least 85%, even more preferably of at least 90% and most preferably of at least 95% or even 97%, with any one of the (G/C modified) NA sequences defined in the fifth column (column "C") of Table I, or of a fragment or variant of any one of these sequences.
  • the at least one coding sequence of the nucleic acid sequence according to the invention comprises or consists of an nucleic acid sequence identical to or having a sequence identity of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
  • RNA sequences defined in the fifth column (column "C") of Table I, or of a fragment or variant of any one of these sequences.
  • the at least one coding sequence of the RNA sequence according to the invention comprises or consists of an nucleic acid sequence having a sequence identity of at least 80% with any one of the (human codon usage adapted) RNA sequences defined in the fifth column (column "C") of Tables I, or of a fragment or variant of any one of these sequences.
  • the present invention provides an nucleic acid sequence as defined herein comprising at least one coding sequence encoding at least one Norovirus peptide or protein derived from Norovirus capsid protein VPI, wherein the coding sequence comprises or consists of any one of the (human codon usage adapted) RNA sequences defined in the fifth column (column "C") of Table I, or of a fragment or variant of any one of these sequences.
  • a nucleic acid molecule of the invention is at least 15, 20, 25 or 30 nucleotides in length. Preferably, it hybridizes under stringent conditions to a nucleic acid molecule comprising a nucleotide sequence of the nucleic acid molecule of the present invention or used in the methods of the present invention, e.g.
  • nucleic acid molecule comprising the sequence shown in SEQ 10 NOs: 4411-39690, 39713-39746.
  • the nucleic acid molecule is preferably at least 20. 30, 50, 100, 250 or more nucleotides in length.
  • a complement sequence derived from SEQ 10 NDs: 4411-39690, 39713-39746, or a fragment or variant thereof is used in the hybridization.
  • a complement of a nucleic acid molecule of the invention that hybridizes under stringent conditions to a sequence shown in SEQ ID NDs: 4411-39690. 39713-39746 correspond to a naturally-occurring nucleic acid molecule of the invention.
  • a "naturally-occurring" nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein).
  • the at least one coding sequence of the nucleic acid sequence according to the invention comprises or consists of an nucleic acid sequence identical to or having a sequence identity of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%. 80%, 85%, 86%, 87%, 88%, 89%, 90%.
  • a "fragment" of an amino acid sequence such as a polypeptide or a protein, e.g. the at least one Norovirus protein as described herein, may typically comprise a sequeoce of a protein or peptide as defined herein, which is, with regard to its amino acid sequence (or the respective coding nucleic acid molecule), N-terminally and/or C- terminally truncated compared to the amino acid sequence of the original (native) protein (or respective coding nucleic acid molecule). Such truncation may thus occur either on the amino acid level or correspondingly on the nucleic acid level.
  • a sequence identity with respect to such a fragment as defined herein may therefore preferably refer to the entire protein or peptide as defined herein or to the entire (coding) nucleic acid molecule of such a protein or peptide.
  • a fragment of an amino acid sequence comprises or consists of a continuous stretch of amino acid residues corresponding to a continuous stretch of amino acid residues in the protein the fragment is derived from, which represents at least 5%, 10%, 20%, preferably at least 30%, more preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, even more preferably at least 70%, and most preferably at least 8D% of the total (i.e. full-length) protein, from which the fragment is derived.
  • a fragment of a protein or of a peptide may furthermore comprise a sequence of a protein or peptide as defined herein, which has a length of for example at least 5 amino acids, preferably a length of at least 6 amino acids, preferably at least 7 amino acids, more preferably at least 8 amino acids, even more preferably at least 9 amino acids: even more preferably at least 10 amino acids: even more preferably at least II amino acids: even more preferably at least 12 amino acids: even more preferably at least 13 amino acids: even more preferably at least 14 amino acids: even more preferably at least 15 amino acids: even more preferably at least 16 amino acids: even more preferably at least 17 amino acids: even more preferably at least 18 amino acids; even more preferably at least 19 amino acids; even more preferably at least 20 amino acids; even more preferably at least 25 amino acids; even more preferably at least 30 amino acids; even more preferably at least 35 amino acids; even more preferably at least 50 amino acids; or most preferably at least ID0 amino acids
  • such fragment may have a length of about 6 to about 2D or even more amino acids, e.g. fragments as processed and presented by MHC class I molecules, preferably having a length of about 8 to about ID amino acids, e.g. 8, 9, or ID, (or even B, 7, II, or 12 amino acids), or fragments as processed and presented by MHC class II molecules, preferably having a length of about 13 or more amino acids, e.g.13, 14, 15, IB, 17, 18, 19, 2D or even more amino acids, wherein these fragments may be selected from any part of 5 the amino acid sequence.
  • These fragments are typically recognized by T-cells in form of a complex consisting of the peptide fragment and an MHC molecule, i.e.
  • fragments are typically not recognized in their native form. Fragments of proteins or peptides may comprise at least one epitape of those proteins or peptides. Furthermore also domains of a protein, like the extracellular domain, the intracellular domain or the transmembrane domain and shortened or truncated versions of a protein may be understood to comprise a fragment of a protein.
  • a "variant" of a protein or a peptide may be generated, having an amino acid sequence, which differs from the original sequence in one or more mutation(s), such as one or more substituted, inserted and/or deleted amino acid(s).
  • these fragments and/or variants have the same biological function or specific activity compared to the full-length native protein, e.g. its specific antigenic property.
  • "Variants" of proteins or peptides as defined in the context of the present5 invention may comprise conservative amino acid substitution(s) compared to their native, i.e. non-mutated physiological, sequence. Those amino acid sequences as well as their encoding nucleotide sequences in particular fall under the term variants as defined herein.
  • amino acids which originate from the same class, are exchanged for one another are called conservative substitutions.
  • these are amino acids having aliphatic side chains, positively or negatively charged side chains, aromatic groups in the side chains or amino acids, the side chains of which can enter into hydrogen bridges,D e.g. side chains which have a hydroxyl function.
  • an amino acid having a polar side chain is replaced by another amino acid having a likewise polar side chain, or, for example, an amino acid characterized by a hydrophobic side chain is substituted by another amino acid having a likewise hydrophobic side chain (e.g.
  • Insertions and substitutions are possible, in particular, at those sequence positions which cause no modification to the three-dimensional structure or do not affect the binding region. Modifications to a three-5 dimensional structure by insertion(s) or deletion(s) can easily be determined e.g. using CD spectra (circular dichroism spectra) (Urry, 1985, Absorption, Circular Dichroism and D D of Polypeptides, in: Modern Physical Methods in Biochemistry, Neuberger et al. (ed.), Elsevier, Amsterdam).
  • a "variant" of a protein or peptide may have at least 50%, 51%, 52%, 53%, 54%, 55%,D 5B%, 57%, 58%, 59%, B0%, 61%. B2%, 63%, 64%. 65%. 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%.
  • a "variant" of a protein or peptide as used herein is at least 40%, preferably at least 50%, more preferably at least 60%, more preferably at least 70%, even more preferably at least 80%, even more preferably5 at least 90%.
  • a protein of the invention refers to any polypeptide being identical or having in increasing order of preference at least 50%, 51%, 52%, 53%, 54%, 55%, 56%. 57%, 58%, 59%, BD%, Gl%, 62%. 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%. 74%, 75%, 76%, 77%, 78%, 79%, 80%.
  • variants of proteins or peptides as defined herein, which may be encoded by a nucleic acid may also comprise those sequences, wherein nucleotides of the encoding nucleic acid sequence are exchanged according to the degeneration of the genetic code, without leading to an alteration of the respective amino acid sequence of the protein or peptide, i.e. the amino acid sequence or at least part thereof may not differ from the original sequence in one or more mutatioo(s) within the above meaning.
  • the at least one coding region of the inventive artificial nucleic acid comprises or consists of at least one nucleic acid sequence according to any one of SE0 ID NOs: 4411-39690, 39713-39746, or a fragment or variant of any of these sequences.
  • a "fragment" of a nucleic acid sequence comprises or consists of a continuous stretch of nucleotides corresponding to a continuous stretch of nucleotides in the full-length nucleic acid sequence which is the basis for the nucleic acid sequence of the fragment, which represents at least 20%. preferably at least 30%. more preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, even more preferably at least 70%, even more preferably at least 80%, and most preferably at least 90% of the full-length nucleic acid sequence.
  • Such a fragment in the sense of the present invention, is preferably a functional fragment of the full-length nucleic acid sequence.
  • a variant of a nucleic acid sequence typically relates to a variant of a nucleic acid sequence, which forms the basis of a nucleic acid sequence.
  • a variant nucleic acid sequence may exhibit one or more nucleotide deletions, insertions, additions and/or substitutions compared to the nucleic acid sequence, from which the variant is derived.
  • a variant of a nucleic acid sequence is at least 40%, preferably at least 50%. more preferably at least 60%, more preferably at least 70%, even more preferably at least 80%, even more preferably at least 90%, most preferably at least 95% identical to the nucleic acid sequence the variant is derived from.
  • the variant is a functional variant.
  • a "variant" of a nucleic acid sequence may have at least 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%. 66%, 67%, 68%.
  • nucleotide identity over a stretch of at least 10, at least 20, at least 30, at least 50, at least 75 or at least 100 nucleotides of such nucleic acid sequence.
  • nuclBic acid sequence encoding a protein of the invention, a capsid protein, a Norovirus capsid protein VPI or a Norovirus capsid protein VP2 as defined herein refers to any nucleic acid sequence having in increasing order of preference at least 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%.
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises or consists of Norovirus capsid protein VPI, or a fragment or variant thereof. More preferably, the at least one encoded polypeptide comprises or consists of an amino acid sequence according to any one of SED ID NOs: 1-4410, or a fragment or variant of any of these sequences.
  • a nucleic acid sequence encoding a Norovirus capsid protein VPI as defined herein refers to any nucleic acid sequence being identical or having in increasing order of preference at least 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%. 59%, 60%, 61%, 62%, 63%, 64%, 65%. 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%.74%.75%, 76%, 77%, 78%, 79%, 80%. 81%. 82%.
  • the at least one encoded polypeptide comprises or consists of a fragment of Norovirus capsid protein VPI or a variant of such a fragment.
  • the at least one encoded polypeptide comprises or consists of a C-terminal fragment of Norovirus capsid protein VPI, or a variant of such a fragment.
  • the at least one encoded polypeptide comprises Dr consists of an N-terminal fragment of Norovirus capsid protein VPI, or a variant of such a fragment.
  • the at least one encoded polypeptide comprises or consists of a fragment, preferably a C-terminal fragment, or a variant of such a fragment, of a Norovirus capsid protein VPI as present in a Norovirus polyprotein (precursor protein) before cleavage.
  • Nonovirus capsid protein VPI as present in a Norovirus polyprotein before cleavage typically refers to a continuous amino acid sequence beginning at the N-terminus of a Norovirus polyprotein (before cleavage) and comprising the amino acid residue immediately N-terminal of the first amino acid residue of a precursor of Norovirus pr protein as present in the Norovirus polyprotein.
  • Non-virus capsid protein VPI as present in a Norovirus polyprotein before cleavage may refer to a part of a Norovirus polyprotein corresponding to Norovirus capsid protein VPI comprising a C-terminal fragment, preferably a C-terminal signal sequence, which is typically not present in mature Norovirus protein VPI.
  • a 'Norovirus capsid protein VPI as present in a Norovirus polyprotein before cleavage' as used herein may comprise an amino acid sequence derived from an amino acid sequence corresponding to amino acid residues I to 122 of a Norovirus polyprotein before cleavage.
  • a Norovirus capsid protein VPI as present in a Norovirus polyprotein before cleavage comprises an amino acid sequence according to any one of SEQ ID NDs: I- 4410, or a fragment or variant of any of these sequences.
  • a "C-terminal fragment, or a variant of such a fragment, of Norovirus capsid protein VPI as present in a Norovirus 5 pnlyprotein (precursor protein) before cleavage” preferably comprises an amino acid sequence corresponding to a continuous amino acid sequence, which is located immediately N-terminal of Norovirus pr protein in a Norovirus polyprotein before cleavage, or to a fragment or variant of said amino acid sequence.
  • the C-terminal fragment, or a variant of such a fragment, of Norovirus capsid protein VPI as present in a Nnrovirus polyprotein (precursor protein) before cleavage comprises or consists of at least 3, 4, 5, G, 7, 8, 9, or, most preferably, at least ID amino acid residues.
  • the C-terminal fragment, nr a ID variant of such a fragment, of Norovirus capsid protein VPI as present in a Norovirus polyprotein (precursor protein) before cleavage may consist of 3 to 4D, 3 to 3D, 3 to 2D, 5 to 2D or ID to 2D amino acid residues.
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises or consists of at least one amino acid sequence derived from a signal sequence, or a fragment 15 Dr variant thereof.
  • signal sequence preferably refers to an amino acid sequence, which is involved in the targeting of a protein, e.g. a Norovirus protein, to a cellular compartment, preferably a membrane, more preferably a membrane of the endoplasmic reticulum (E ).
  • a signal sequence in the context of the present invention preferably comprises from 3 to 4D, 3 to 0 3D, 3 to 2D, 5 to 20 or 10 to 2D amino acid residues.
  • Such a signal sequence may be present, for example, in a Norovirus polyprotein and may be removed during processing of said polyprotein.
  • a signal sequence is preferably no longer present in a mature Norovirus protein.
  • Norovirus capsid protein VPI as present in a Norovirus polyprotein typically comprises a C-terminal signal sequence, corresponding to the amino acid sequence immediately N-terminal of Norovirus pr protein (e.g. amino acid residues ID5 to 122 in a Nnrovirus polyprotein before cleavage). That signal sequence is involved in targeting Norovirus 5 capsid protein VPI to the ER membrane and is typically removed in order to yield mature Norovirus capsid protein VPI, which no longer comprises said C-terminal fragment comprising a signal sequence.
  • the amino acid sequence derived from a signal sequence, or a fragment or variant thereof comprises at least 3, 4, 5, 6, 7, 8, 3, ID, II, 12, 13, 14, 15, IB, 17, 18, 19 or at least 2D amino acid residues.
  • the amino acid sequence derived from D a signal sequence, or a fragment or variant thereof may consist of 3 to 4D, 3 to 30, 3 to 2D, 5 to 20 or ID to 2D amino acid residues.
  • the amino acid sequence derived from a signal sequence, or a fragment or variant thereof consists of from 3 to 2D amino acid residues.
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial 35 nucleic acid comprises or consists of at least one amino acid sequence derived from a signal sequence, which comprises or consists of an amino acid sequence that is bound by signal recognition particle (SRP). More preferably, the at least one amino acid sequence derived from a signal sequence comprises or consists of an amino acid sequence that is recognized by signal peptide peptidase (SPP), by a viral protease and/or by furin or a furin-like protease.
  • SRP signal recognition particle
  • the at least one amino acid sequence derived from a signal sequence comprises an amino acid sequence that is recognized by a viral protease comprising one or more of a Norovirus non-structural protein selected from the group consisting of NSI/NS2, NS3, NS4, NS5, NSB. and NS7.
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises or consists of at least one amino acid sequence derived from a signal sequence of a secretory protein or from a signal sequence of a membrane protein. More preferably, the at least one amino acid sequence derived from a signal sequence, preferably derived from a signal sequence of a membrane protein, targets the at least one encoded protein to a ID cellular compartment, preferably to the endoplasmic reticulum (E ), more preferably to the ER membrane.
  • E endoplasmic reticulum
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises or consists of an amino acid sequence corresponding tc a signal sequence from a Norovirus protein, preferably from Norovirus capsid protein VPI, more preferably from Norovirus capsid protein VPI as present in a Norovirus polyprotein 15 before cleavage, or a fragment or variant of any of these.
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises or consists of an amino acid sequence corresponding to a signal sequence from Norovirus capsid protein VPI as present in a Norovirus polyprotein before cleavage, or a fragment or variant thereof, wherein the signalD sequence is preferably derived from a C-terminal fragment of Norovirus capsid protein VPI as present in a Norovirus polyprotein before cleavage, preferably as described herein.
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises or consists of a fragment, preferably a C-terminal fragment, or a variant of such a5 fragment, of a mature Norovirus protein, preferably of a mature Norovirus capsid protein VPI.
  • the mature Norovirus protein is a mature Norovirus protein as defined herein.
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises or consists of a fragment, preferably a C-terminal fragment, or a variant of such a fragment, ofD mature Norovirus capsid protein VPI, wherein the mature Norovirus capsid protein VPI does preferably not comprise a C-terminal signal sequence as described herein with respect to a Norovirus capsid protein VPI as present in a Norovirus polyprotein (before cleavage). More preferably, the mature Norovirus capsid protein VPI comprises or consists of an amino acid sequence according to any one of SEQ ID NOs: 1-4410, or a fragment or variant thereof.
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises or consists of a C-terminal fragment, preferably as defined herein, or a variant of such a fragment, of mature Norovirus capsid protein VPI.
  • the C-terminal fragment, or a variant of such a fragment, of mature Norovirus capsid protein VPI comprises or consists of at least 3, , 5, 6, 7. 8, 9, or, most preferably, at least ID amino acid residues.
  • the C-terminal fragment, or a variant of such a fragment, of mature Norovirus capsid protein VPI may comprise or consist of 3 to 40, 3 to 3D, 3 to 2D, 3 to ID, 5 to 20 or 10 to 20 amino acid residues.
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises or consists of an amino acid sequence derived from a
  • amino acid sequence according to a) may be in continuation with the amino acid sequence according to b), whereinD the sequences may be positioned relative to each other in any manner.
  • amino acid sequences according to a) and b) may be separated in the at least one encoded protein by another amino acid sequence.
  • amino acid sequence according to a) is located N-terminally with respect to b).
  • the at least Dne polypeptide encoded by the at least one coding region of the inventive5 artificial nucleic acid comprises or consists of at least one amino acid sequence corresponding to a fragment of Norovirus nonstructural protein I (NSI/2), or a variant of such a fragment.
  • fragment of Norovirus non-structural protein I preferably relates to a continuous amino acid sequence derived from Norovirus non-structural protein I (NSI/2), or to a fragment or variant of said continuous amino acidD sequence.
  • the fragment, or variant thereof, of Norovirus non-structural protein I comprises or consists of at least 3, 4, 5, G, 7, 8, 9, or, most preferably, at least 10 amino acid residues.
  • the fragment, or variant thereof, of Norovirus non-structural protein I may comprise or consist of 3 to 40, 3 to 30. 3 to 20, 3 to 10, 5 to 2D or 10 to 2D amioo acid5 residues.
  • the fragment, or variant thereof, of Norovirus non-structural protein I (NSI/2) comprises or consists of from 3 to 20 amino acid residues.
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises or consists of at least one amino acid sequence corresponding to an N-terminal fragment of Norovirus non-structural protein I (NSI/2). or a variant of said fragment.
  • N-terminal fragment of Norovirus non-structural protein I relates to a continuous amino acid sequence derived from the N-tBrminus of Norovirus non-structural protein I (NSI/2). More preferably, the N-terminal fragment of Norovirus non-structural protein I (NSI/2) comprises or consists of from 3 to 2D amino acid residues.
  • the at least one encoded polypeptide comprises an N-terminal fragment of Norovirus non-structural I0 protein I (NSI/2), wherein the N-terminal fragment of Norovirus non-structural protein I (NSI/2) is a continuous amino acid sequence comprising or consisting of 3 to 2D amino acid residues corresponding to a continuous amino acid sequence of 3 to 2D amino acid residues in the first 2D amino acid residues (counting from the N-terminus) of Norovirus non-structural protein I (NSI/2), or a variant thereof.
  • the at least one encoded polypeptide comprises or consists of an N-terminal fragment of Norovirus non ⁇ structural protein I (NSI/2), wherein the N-terminal fragment of Norovirus non-structural protein I (NSI/2) is a continuous amino acid sequence comprising or consisting of 3 to 20 amino acid residues corresponding to a continuous amino acid sequence of 3 to 2D amino acid residues in the first 2D amino acid residues (counting from the N-terminus) of a mature Norovirus non ⁇ structural protein I (NSI/2), or a variant thereof.
  • the first 2D amino acid residoes of a mature Norovirus non-structuralD protein I preferably comprise or consist of the N-terminus itself (i.e. the amino acid residue at the N-terminus) and the I9 following amino acid residues.
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises a first Norovirus protein, which is preferably a Norovirus protein as described herein, or a fragment or5 variant thereof, and further comprises at least one second or further Norovirus protein, or a fragment or variant thereof, wherein the at least one second or further Norovirus protein, or the fragment or variant thereof, is distinct from the first Norovirus protein, or the fragment or variant thereof.
  • the first Norovirus protein is preferably selected from the groop consisting of Norovirus NSI/NS2, NS32A,0 NS42B, NS53, NSG4A, NS4B or NS75, or a fragment or variant thereof.
  • the second or further Norovirus protein is selected from the group consisting of Norovirus capsid protein VPI, Norovirus capsid protein VP2 and a Norovirus non-structural protein, preferably Norovirus NSI/NS2, NS3, NS4, NS5, NSB, or NS7, or a fragment or variant thereof.
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid5 comprises Norovirus capsid protein VPI and/or VP2, or a fragment or variant thereof, and further comprises at least one of the following: a) an amino acid sequEncE corrEsponding to a C-terminal fragment, or a variant thereof, of mature Norovirus capsid protein VPI, preferably as described herein;
  • N-terminal fragment an amino acid sequence corresponding to an N-terminal fragment, or a variant thereof, of Norovirus non-structural protein I (NSI/NS2), preferably as described herein; and/or
  • the at least one polypeptide encoded by the at least one coding region of the inventive artificial nucleic acid comprises or consists of, preferably in this order from N-terminus to C-terminus, Norovirus capsid protein VPI, or a fragment or variant thereof,
  • N-NS2 Norovirus non-structural protein I
  • the inventive artificial nucleic acid is monocistronic, bicistronic or multicistronic.
  • the inventive artificial nucleic acid is monocistronic.
  • the inventive artificial nucleic acid comprises one coding region, wherein the coding region encodes a polypeptide comprising at least two different Norovirus proteins, preferably as defined herein, or a fragment or variant thereof.
  • the inventive artificial nucleic acid can be bi- or multicistronic and comprises at least two coding regions, wherein the at least two coding regions encode at least two polypeptides, wherein each of the at least two polypeptides comprises at least one different Norovirus protein, preferably as described herein, or a fragment or variant of any one of these proteins.
  • the inventive artificial nucleic acid may comprise two coding regions, wherein the first ending region encodes a first polypeptide comprising a first Norovirus protein, or a fragment or variant thereof, and wherein the second coding region encodes a second polypeptide comprising a second Norovirus protein, or a fragment or variant thereof, wherein the first and second Nnrovirus proteins or a fragment or variant thereof are distinct from each other.
  • the inventive artificial nucleic acid may be provided as DNA or as NA, preferably an RNA as defined herein. More preferably, the inventive artificial nucleic acid is an artificial mRNA.
  • the inventive artificial nucleic acid may further be single stranded or double stranded.
  • the inventive artificial nucleic acid preferably comprises a sense and a corresponding antisense strand.
  • the inventive artificial nucleic acid as defined herein typically comprises a length of about 50 to about 20000, or 100 to about 20000 nucleotides, preferably of about 25D to about 20000 nucleotides, more preferably of about 500 to about 10000, even more preferably of about 500 to about 5000.
  • the inventive artificial nucleic acid as defined herein may be in the form of a modified nucleic acid, preferably a modified mRNA, wherein any modification, as defined herein, may be introduced into the inventive artificial nucleic 5 acid. Modifications as defined herein preferably lead to a stabilized artificial nucleic acid, preferably a stabilized artificial RNA, of the present invention.
  • the inventive artificial nucleic acid preferably an mRNA
  • a stabilized nucleic acid preferably as a “stabilized mRNA”
  • a nucleic acid preferably an mRNA, that is essentially resistant
  • a backbone modification in connection with the present invention is a modification in which phosphates of the backbone of the nucleotides contained in the mRNA are chemically modified.
  • Nucleotides that may be preferably used in this connection contain e.g. a phosphorothioate-modified phosphate backbone, preferably at least one of the phosphate oxygens contained in the phosphate backbone being replaced by a sulfur atom.
  • 15 acids may further include, for example: non-ionic phosphate analogues, such as, for example, alkyl and aryl phosphonates, in which the charged phosphonate oxygen is replaced by an alkyl or aryl group, or phosphodiesters and alkylphosphotriesters, in which the charged oxygen residue is present in alkylated form.
  • non-ionic phosphate analogues such as, for example, alkyl and aryl phosphonates, in which the charged phosphonate oxygen is replaced by an alkyl or aryl group
  • phosphodiesters and alkylphosphotriesters in which the charged oxygen residue is present in alkylated form.
  • backbone modifications typically include, without implying any limitation, modifications from the group consisting of methylphosphonates, phosphoramidates and phosphorothioates (e.g. cytidine-5'-0-(l-thiophosphate)).
  • nucleic acid modification may refer to chemical modifications comprising backbone modifications as well as sugar modifications or base modifications.
  • a modified artificial nucleic acid preferably an mRNA, as defined herein may contain nucleotide analogues/modifications, e.g. backbone modifications, sugar modifications or base modifications.
  • a backbone modification in connection with the present invention is a modification, in which phosphates of the backbone of the nucleotides contained in anD artificial nucleic acid, preferably an mRNA, as defined herein are chemically modified.
  • a sugar modification in connection with the present invention is a chemical modification of the sugar of the nucleotides of the artificial nucleic acid, preferably an mRNA, as defined herein.
  • a base modification in connection with the present invention is a chemical modification of the base moiety of the nucleotides of the artificial nucleic acid, preferably an mRNA.
  • nucleotide analogues or modifications are preferably selected from nucleotide analogues, which are applicable for transcription and/ or translation. 5 Sugar Modifications:
  • modified nucleosides and nucleotides which may be incorporated into a modified artificial nucleic acid, preferably an m NA, as described herein, can be modified in the sugar moiety.
  • the 2' hydroxyl group (OH) can be modified or replaced with a number of different "oxy" or “deoxy” substituents.
  • Examples of "oxy" -2' hydroxy! group modifications include, but are not 5 limited to, alkoxy or aryloxy (-DR.
  • R H, alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar); polyethyleneglycols (PEG), - D(CH2CHzQ)nCH2CH2DR; "locked" nucleic acids (LNA) in which the 2' hydroxyl is connected, e.g., by a methylene bridge, to the 4' carbon of the same ribose sugar; and amino groups (-D-amino, wherein the amino group, e.g., HRR, can be alkylamino, dialkylamino, heteracyclyl. arylamino, diarylaminn, heteroarylamino, or diheteroaryl amino, ethylene diamine, polyamino) or aminoalkoxy.
  • Deoxy modifications include hydrogen, amino (e.g. NH2; alkylamino, dialkylamino, heteracyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid); or the amino group can be attached to the sugar through a linker, wherein the linker comprises one or more of the atoms C, N, and D.
  • amino e.g. NH2; alkylamino, dialkylamino, heteracyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid
  • the sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose.
  • an artificial nucleic acid preferably an mRNA, can include nucleotides containing, for 15 instance, arabinose as the sugar.
  • the phosphate backbone may further be modified in the modified nucleosides and nucleotides, which may be incorporated into a modified artificial nucleic acid, preferably an mRNA, as described herein.
  • the phosphate groups of the backbone can be modifiedD by replacing one or more of the oxygen atoms with a different substituent.
  • the modified nucleosides and nucleotides can include the full replacement of an unmodified phosphate moiety with a modified phosphate as described herein.
  • modified phosphate groups include, but are not limited to, phosphorothioate, phosphoroselenates, borano phosphates, borano phosphate esters, hydrogen phosphonates, phosphoroamidates, alkyl or aryl phosphonates and phasphotriesters.
  • Phosphorodithiaates have both non-linking oxygens replaced by sulfur.
  • the phosphate linker can also be modified by the5 replacement of a linking oxygen with nitrogen (bridged phosphoroamidates), sulfur (bridged phosphorDthioates) and carbon (bridged methylene-phosphonates).
  • modified nucleosides and nucleotides which may be incorporated into a modified nucleic acid, preferably an mRNA, asD described herein can further be modified in the nucleobase moiety.
  • nuclenbases found in a nucleic acid such as RNA include, but are not limited to. adenine, guanine, cytosine and uracil.
  • the nucleosides and nucleotides described herein can be chemically modified on the major groove face.
  • the major groove chemical modifications can include an amino group, a thiol group, an alkyl group, or a halo group.
  • the nucleotide analogues/ modifications are selected from base modifications, which are preferably selected from 2-amino-B-chloropurineriboside-5'-triphosphate, 2-Aminopurine-riboside-5'- triphasphate; 2-aminaadenosine-5'-triphosphatE, 2'-Amino-2'-deoxycytidine-triphDsphatB, 2-thiocytidinB-5'-triphosphate.
  • 2- thiouridinB-5'-triphosphatE 2- thiouridinB-5'-triphosphatE, 2'-FluarathymidinB-5'-triphosphate, 2'-D-Methyl inDsine-5'-triphosphate 4-thiouridine-5'- triphosphate, 5-aminoallylcytidine-5'-triphasphatB, 5-aminoallyluridine-5'-triphosphatB, 5-brDmacytidinB-5'-triphDsphatE, 5- bromouridine-5'-triphosphate, 5-Bromo-2'-deoxycytidine-5'-triphosphate, 5-BromQ-2'-dEDxyuridine-5'-triphosphate, 5- iodocytidinE-5'-triphasphatE, 5-lado-2'-dEoxycytidinE-5'-triphosphatB.
  • 5-iodouridinE-5'-triphosphatB 5-lodo-2'-dBoxyuridine- 5'-triphosphate, 5-mBthylcytidinE-5'-triphDsphatE, 5-mBthyluridinE-5'-triphosphate, 5-PropynyI-2'-dBOxycytidine-5'- triphosphate, 5-Prapynyl-2'-dEDxyuridinE-5'-triphDsphatE, B-azacytidine-S'-triphosphate, B-azauridine-5'-triphosphatB, 6- chlDrapurinEribDside-5'-triphosphatB, 7-deazaadenDsine-5'-triphDsp ate, 7-dBazaguanasinB-5'-triphosphatB, 8-azaadenasine- 5'-triphosphatB.
  • nuclBotidEs for bass modifications sElsctEd from the group of basB-modifiEd nuclBotidEs consisting of 5-methylcytidine-5'- triphosphatB, 7-dEazaguanosinB-5'-triphosphatB, 5-bromocytidine-5'-triphosphate. and pseudouridine-5'-triphosphatE.
  • modified nuclBosides includedE pyridin-4-nnB ribonuclBosidE. 5-aza-uridine, 2-thio-5-aza-uridine, 2- thiouridine, 4-thio-pseudouridine, 2-thio-psBudouridinB, 5-hydroxyuridinB, 3-mBthyluridine, 5-carboxymBthyl-uridine, I- carboxymethyl-psEudouridinB, 5-propynyl-uridine, 1-propynyl-psEudouridinE, 5-taurinomethyluridinB.
  • modified nucleosidBs includedE 5-aza-cytidinB, pseudoisocytidinE, 3-mBthyl-cytidinB, N4-acetylcytidinE, 5- formylcytidinE, N4-methylcytidine, 5-hydroxymethylcytidinE, 1-mEthyl-psEudoisocytidinE, pyrrolo-cytidinE, pyrrolo- psBudoisocytidinE, 2-thio-cytidinE, 2-thio-5-mEthyl-cytidine, 4-thio-pseudoisocytidine, 4-thio-l-mBthyl-pseudoisocytidine, 4-thio- l-mEthyl-l-deaza-psBudoisocytidinE, 1-methyl-l-dEaza-p
  • modified nucleosides include inosine, 1-mBthyl-inosinE, wyosinE, wybutnsine, 7-dsaza-guanosinE, 7-deaza-
  • the nucleotide can be modified on the major groove face and can include replacing hydrogen on C-5 of uracil with a methyl group or a halo group.
  • a modified nucleoside is 5'-D-(l-thiophusphatE)-adenosine, 5'-0-(l-thiophosphate)-cytidine, 5'-0-(l-thiophosphate)-guanosine, 5'-0-(l-thiophosphate)-uridine or 5'-0-(l-thiophosphate)- pseudouridine.
  • a modified artificial nucleic acid may comprise nucleoside modifications selected from 6-aza-cytidine, 2-thiD-cytidine, a-thio-cytidine, Pseudo-isu-cytidine, 5-aminoaHyl-uridine, 5-iodo-uridinB, Nl- methyl-pseudouridine, 5,6-dihydrouridine, a-thio-uridine, 4-thio-uridine, B-aza-uridine, 5-hydroxy-uridine, deoxy-thymidine, 5- methyl-uridine, Pyrrolo-cytidine, inosine, a-thio-guanasine, B-methyl-guanosine, 5-methyl-cytdine, 8-oxo-guanosinE, 7-deaza- guanosine, Nl-methyl-adenosine, 2-amino-B-Chloro-pur
  • a modified artificial nucleic acid preferably an mRNA, as defined herein can contain a lipid modification.
  • a lipid-modified artificial nucleic acid as defined herein typically further comprises at least one linker
  • the lipid-modified artificial nucleic acid comprises at least one artificial nucleic acid as defined herein and at least one (Afunctional) lipid covalently linked (without a linker) with that artificial nucleic acid.
  • the lipid- modified artificial nucleic acid comprises an artificial nucleic acid molecule as defined herein, at least Dne linker covalently linked with that artificial nucleic acid, and at least one lipid covalently linked with the respective linker, and also at least oneD (Afunctional) lipid covalently linked (without a linker) with that artificial nucleic acid.
  • the lipid modification is present at the terminal ends of a linear artificial nucleic acid.
  • the artificial nucleic acid of the present invention may be modified, and thus stabilized, by modifying the G/C content of the artificial nucleic acid, preferably an mRNA, preferably of the coding region of the inventive artificial nucleic acid.
  • the G/C content of the at least one coding region of the artificial nucleic acid is modified,D preferably increased, compared to the G/C content of the corresponding coding sequence of the wild type nucleic acid, preferably an mRNA, wherein the encoded amino acid sequence is preferably not modified compared to the amino acid sequence encoded by the corresponding wild type nucleic acid (i.e. the non-modified nucleic acid), preferably an mRNA.
  • This modification of the inventive artificial nucleic acid, preferably of an mRNA, as described herein is based on the fact that the sequence of any mRNA region to be translated is important for efficient translation of that mRNA.
  • the composition and the sequence of various 5 nucleotides are important.
  • sequences having an increased G (guanosine)/C (cytosine) content are more stable than sequences having an increased A (adenosine)/U (uracil) content.
  • the codons of the artificial nucleic acid preferably an mRNA
  • the codons of the artificial nucleic acid are therefore varied compared to the respective wild type mRNA, while retaining the translated amino acid sequence, such that they include an increased amount of G/C nucleotides.
  • the most favorable codons for the stability can be determined (so-called alternative codon usage).
  • the amino acid to be encoded by the artificial nucleic acid 5 preferably an mRNA
  • amino acids which are encoded by codons which contain exclusively G or C nucleotides
  • no modification of the codon is necessary.
  • the codons for Pro CCC or CCG
  • Arg CGC or CGG
  • Ala GCC or GCG
  • GGC or GGG require no modification, since no A or U is present.
  • codons which contain A and/or U nucleotides can be modified by substitution of other codons, which code for the same amino acids but contain no A and/or U. Examples of these are: the codons for Pro can be modified by substitution of other codons, which code for the same amino acids but contain no A and/or U. Examples of these are: the codons for Pro can be modified by substitution of other codons, which code for the same amino acids but contain no A and/or U. Examples of these are: the codons for
  • the codons for Arg can be modified from CGU or CGA or AGA or AGG to CGC or CGG; the codons for Ala can be modified from GCU or GCA to GCC or GCG; the codons for Gly can be modified from GGU or GGA to GGC or GGG.
  • a or U nucleotides cannot be eliminated from the codons, it is however possible to decrease the A and U content by using codons, which contain a lower content of A and/ or U nucleotides. Examples of these are: the codons for Phe can be modified from UUU to UUC; the codons for Leu can be modified from UUA, UUG, CUU or CUA to CUC or CUG; the codons
  • the codon for Ser can be modified from UCU or UCA or AGU to UCC, UCG or AGC; the codon for Tyr can be modified from UAU to UAC; the codon for Cys can be modified from UGU to UGC; the codon for His can be modified from CAU to CAC; the codon for Gin can be modified from CAA to CAG: the codons for He can be modified from AUU or AUA to AUG; the codons for Thr can be modified from ACU or ACA to ACC or ACG; the codon for Asn can be modified from AAU to AAC; the codon for Lys can be modified from AAA to AAG; the codons for Val can be modified from GUU or GUA to GUC or GUG; the codon for Asp can be modified from GAU to GAC;0 the codon for Glu can be modified from GAA to GAG; the stop codon UAA can be modified to UAG or UGA.
  • the G/C content of the coding region of the inventive artificial nucleic acid is increased by at least 7%. more preferably by at least 15%, particularly preferably by at least 20%, compared to the G/C content of the coding region of the wild type nucleic acid.
  • the G/C content of the inventive artificial nucleic acid to the maximum (i.e.100% of the substitutable codons), in particular in the region coding for the at least one protein, compared to the wild type sequence.
  • the G/C content of the artificial nucleic acid of the invention is increased compared to the G/C content of the corresponding coding sequence of the wild type mRNA, or wherein the C content of the coding region of the mRNA sequence is increased compared to the C content of the corresponding coding sequence of the wild type mRNA, or wherein the codon usage in the coding region of the mRNA sequence is adapted to the human codon usage, or wherein the codon adaptation index (CAI) is increased or maximised in the coding region of the mRNA sequence, wherein the encoded amino acid sequence of the mRNA sequence is preferably not being modified compared to the encoded amino acid sequence of the wild type mRNA.
  • CAI codon adaptation index
  • the artificial nucleic acid according to the invention is codon optimized
  • the at least one coding region comprises a nucleic acid sequence, which is codon-optimized; and/ or
  • the at least one coding sequence comprises a nucleic acid sequence, which is identical or at least 50%, BD%, 70%, 80%, 85%, 86%. 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to a nucleic acid sequence selected from the group consisting of SEQ 10 NOs: 8821-13230, 26461-39690, 39715, 39716, 39717, 39720, 39721, 39724, 39725, 39728, 39729, 39730, 39733, 39734, 39737, 39738, 39741, 39742, 39745 and 39746, or a fragment or variant of any of these sequences; and/or
  • the at least one coding sequence comprises a nucleic acid sequence, which is identical or at least 50%, 60%, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 13231-17640, or a fragment or variant of any of these sequences; and/or
  • the artificial nucleic acid of the invention wherein the at least one coding sequence comprises a nucleic acid sequence, which is identical or at least 50%. 60%, 7D%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 17641- 22050, or a fragment or variant of any of these sequences; and/ or
  • the artificial nucleic acid of the invention wherein the at least one coding sequence comprises a nucleic acid sequence, which is identical or at least 50%, 60%, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to a nucleic acid sequence selected from the group consisting of SEQ ID NDs: 22051- 26460, or a fragment or variant of aoy of these sequences.
  • a further preferred modification of the artificial nucleic acid of the present invention is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells.
  • the at least one coding region of the artificial nucleic acid according to the invention comprises a nucleic acid 5 sequence, which is codon-optimized.
  • codon-optimized typically refers to an artificial nucleic acid, preferably to a nucleic acid sequence in the at least one coding region therein, wherein at least one codon of the wild type sequence, which codes for a tRNA which is relatively rare in the cell, is exchanged for a codon, which codes for a tRNA which is relatively frequent in the cell and carries the same amino acid as the relatively rare tRNA.
  • that modification also increases the G/C content of the at least one coding region of the artificial nucleic acid.
  • the corresponding modified nucleic acid sequence preferably an mRNA sequence
  • the corresponding modified nucleic acid sequence is translated to a significantly poorer degree than in the case where codons coding for relatively "frequent" tRNAs are present.
  • the region which encodes the at least one protein as defined herein is modified compared 15 to the corresponding region of the wild type nucleic acid, preferably an mRNA, such that at least one codon of the wild type sequence, which codes for a tRNA which is relatively rare in the cell, is exchanged for a codon, which codes for a tRNA which is relatively frequent in the cell and carries the same amino acid as the relatively rare tRNA.
  • the sequences of the artificial nucleic acid of the present invention is modified such that codons for which frequently occurring tRNAs are available are inserted.
  • the Gly codon, which uses the tRNA, which occurs the most frequently in the (human) cell are particularly5 preferred.
  • This preferred embodiment allows provision of a particularly efficiently translated and stabilized (modified) artificial nucleic acid of the present invention.
  • an artificial nucleic acid of the present invention as described above0 (increased G/C content; exchange of tRNAs) can be carried out using the computer program explained in WD D2/098443 - the disclosure content of which is included in its full scope in the present invention.
  • the nucleotide sequence of any desired mRNA can be modified with the aid of the genetic code or the degenerative nature thereof such that a maximum G/C content results, in combination with the use of codons which code for tRNAs occurring as frequently as possible in the ceil, the amino acid sequence encoded by the artificial nucleic acid preferably not being modified compared to the non- 5 modified sequence.
  • the A/U content in the environment of the ribosome binding site of the artificial nucleic acid of the present invention is increased compared to the A/U content in the environment of the ribosome binding site of its particular wild type nucleic acid, preferably an m NA.
  • This modification increases the efficiency of ribosome binding to the artificial nucleic acid.
  • an effective binding of the ribosomes to the ribosome binding site in turn has the effect of an efficient translation of the artificial nucleic acid.
  • the artificial nucleic acid of the present invention may be modified with respect to potentially destabilizing sequence elements.
  • the coding region and/or the 5' and/or 3' untranslated region of the artificial nucleic acid may be modified compared to the particular wild type nucleic acid such that it contains no destabilizing sequence elements, the amino acid sequence encoded by the modified artificial nucleic acid preferably not being modified compared to its particular wild type nucleic acid.
  • DSE destabilizing sequence elements
  • AU-rich sequences which occur in 3'-UTR sections of numerous unstable RNAs (Caput et al dislike Proc. Natl. Acad. Sci. USA I98B, 83: 1670 to IB74).
  • the artificial nucleic acid of the present invention is therefore preferably modified compared to the wild type nucleic acid such that the artificial nucleic acid contains no such destabilizing sequences.
  • sequence motifs which are recognized by possible endonucleases, e.g. the sequence GAACAAG, which is contained in the 3'-UTR segment of the gene which codes for the transferrin receptor (Binder et al., E BU J. 1394, 13: 19B9 to 1980).
  • sequence motifs are also preferably removed in the artificial nucleic acid of the present invention.
  • the artificial nucleic acid of the present invention has, in a modified form, at least one IRES as defined above and/or at least one 5' and/or 3' stabilizing sequence, in a modified form, e.g. to enhance ribosome binding or to allow expression of different encoded polypeptides located on an artificial nucleic acid of the present invention.
  • the artificial nucleic acid is bi- or multicistronic and wherein an IRES is preferably located between individual coding regions.
  • the at least one coding region of the artificial nucleic acid or artificial nucleic acid molecule comprises or consists of at least one nucleic acid sequence according to any one of SEQ ID NOs: 8821-39690, 39715. 397IB, 39717, 39720, 39721, 39724. 39725, 39728, 39729, 39730, 39733, 39734, 39737, 39738, 39741, 39742, 30745 and 39746, or a fragment or variant of any of these sequences.
  • the at least one coding region of the artificial nucleic comprises or consists of an RNA sequence, which is at least 5D , 51%, 52%, 53%, 54%, 55%, 56%. 57%, 58%, 59%, 60%, 61%. 62%. 63%, 64%, 65%. 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%.
  • an RNA sequence which is at least 5D , 51%, 52%, 53%, 54%, 55%, 56%. 57%, 58%, 59%, 60%, 61%. 62%. 63%, 64%, 65%. 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%
  • the RNA of the present invention may be modified, and thus stabilized, by modifying the guanosine/ cytosine (G/C) content of the RNA, preferably of the at least one coding sequence of the RNA of the present invention.
  • G/C guanosine/ cytosine
  • the G/C content of the coding region of the RNA of the present invention is particularly preferred.
  • ID invention is modified, particularly increased, compared to the G/C content of the coding region of the respective wild type RNA, i.e. the unmodified RNA.
  • the amino acid sequence encoded by the RNA is preferably not modified as compared to the amino acid sequence encoded by the respective wild type RNA.
  • This modification of the RNA of the present invention is based on the fact that the sequence of any RNA region to be translated is important for efficient translation of that RNA.
  • the composition of the RNA and the sequence of various nucleotides are important. In particular, sequences having an increased G (guanosine)/C
  • the codons of the RNA are therefore varied compared to the respective wild type RNA, while retaining the translated amino acid sequence, such that they include an increased amount of G/C nucleotides.
  • the most favourable codons for the stability can be determined (so-called alternative codon usage).
  • the codons for Pro can be modified from CCU or CCA to CCC5 or CCG: the codons for Arg can be modified from CGU or CGA or AGA or AGG to CGC or CGG; the codons for Ala can be modified from GCU or GCA to GCC or GCG; the codons for Gly can be modified from GGU or GGA to GGC or GGG.
  • the codons for Pro can be modified from CCU or CCA to CCC5 or CCG
  • the codons for Arg can be modified from CGU or CGA or AGA or AGG to CGC or CGG
  • the codons for Ala can be modified from GCU or GCA to GCC or GCG
  • the codons for Gly can be modified from GGU or GGA to GGC or GGG.
  • the codons for Phe can be modified from UUU to UUC; the codons for Leu can be modified from UUA. DOG, CUU or CUA to CUC or CUG; the codons for Ser can be modified fromD UGU or UCA or AGU to UCC, UCG or AGC; the codon for Tyr can be modified from UAU to UAC; the codon for Cys can be modified from UGU to UGC; the codoo for His can be modified from CAU to CAC; the codon for Gin can be modified from CAA to CAG; the codons for He can be modified from AUU or AUA to AUG; the codons for Thr can be modified from ACU or ACA to ACC or AGG; the codon for Asn can be modified from AAU to AAC; the codon for Lys can be modified from AAA to AAG; the codons for Val can be modified from GUU or GUA to GUC or BUG; the codon for Asp can be modified from GAU to G
  • the G/C content of the coding region of the RNA of the present invention is increased by at least 7%, more preferably by at least 15%, particularly preferably by at least 2D%, compared to the G/C content of the coding region of the wild type RNA, which codes for an antigen as defined herein or a fragment or variant thereof.
  • the G/C content of the coding region of the wild type RNA which codes for an antigen as defined herein or a fragment or variant thereof.
  • a specific embodiment at least 5%, 10%. 20%, 30%, 40%, 50%, 60%.
  • RNA of the present invention preferably of the at least one coding region of the RNA according to the invention, ta the maximum (i.e. 100% of the substitutable codons) as compared to the wild type sequence.
  • RNA of the present invention is based an the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells.
  • the corresponding modified RNA sequence is translated to a significantly poorer degree than in the case where codons coding for relatively "frequent" tRNAs are present.
  • the region which codes for an antigen as defined herein or a fragment or variant thereof is modified compared to the corresponding region of the wild type RNA such that at least one codon of the wild type sequence, which codes for a tRNA which is relatively rare in the cell, is exchanged far a codon, which codes for a tRNA which is relatively frequent in the cell and carries the same amino acid as the relatively rare tRNA.
  • the sequences of the RNA of the present invention are modified such that codons for which frequently occurring tRNAs are available are inserted.
  • codons of the wild type sequence which code for a tRNA which is relatively rare in the cell, can in each case be exchanged for a codon, which codes for a tRNA which is relatively frequent in the cell and which, in each case, carries the same amino acid as the relatively rare tRNA.
  • Which tRNAs occur relatively frequently in the cell and which, in contrast, occur relatively rarely is known to a person skilled in the art; cf. e.g. Akashi, Curr. Dpin. Genet. Dev. 2DDI, 1KB): GG0-BG6.
  • the codons, which use 5 fur the particular amino acid the tRNA which occurs the most frequently e.g.
  • the Gly codon which uses the tRNA, which occurs the most frequently in the (human) cell, are particularly preferred.
  • This preferred embodiment allows provision of a particularly efficiently translated and stabilized (modified) RNA of the present
  • RNA of the present invention as described above (increased G/C content; exchange of tRNAs) can be carried out using the computer program explained in WD 02/098443 - the disclosure content of which is included in its full scope in the present invention.
  • the nucleotide sequence of any desired RNA can be modified with the aid of the genetic code or the degenerative nature thereof such that a maximum G/C content results, in combination with the use of codons which code for tRNAs occurring as frequently as possible in the cell, the amino acid sequence
  • the modified RNA preferably not being modified compared to the non-modified sequence.
  • the source code in Visual Basic 6.0 development environment used: Microsoft Visual Studio Enterprise G.D with Servicepack 3
  • the A/U content in the environment of the ribosome binding site of the RNA of the present invention is increased compared tD the A/U content in the environment of the ribosome binding site of its
  • RNA of the present invention may be modified with respect to potentially destabilizing sequence elements. Particularly, the coding region and/or the 5' and/or 3' untranslated region of this RNA may be
  • the encoded amino acid sequence of the modified RNA preferably not being modified compared to its respective wild type RNA.
  • DSE destabilizing sequence elements
  • RNA of the present invention is therefore preferably modified compared tu the respective wild type RNA such that the RNA of the
  • 35 present invention contains no such destabilizing sequences. This also applies to those sequence motifs which are recognized by possible endonucleases. e.g. the sequence GAACAAG, which is contained in the 3'-UTR segment of the gene encoding the transferrin receptor (Binder et al., EMBO J. I3B4, 13: 19BB to 1980). These sequence motifs are also preferably removed in the NA of the present invention.
  • the present invention provides an RNA as defined herein comprising at least one coding sequence, wherein the coding sequence comprises or consists of any one of the (modified) nucleic acid sequences defined in SEO ID NOs: 8821-13230. 39715, 3B7IB, 39717, 39720, 39721, 39724, 39725, 33728, 3B729, 39730, 33733, 3B734, 39737, 33738, 33741, 39742, 39745.
  • the at least one coding sequence preferably comprises or consists of a nucleic acid sequence selected from the group consisting of SEQ 10 NOs: 8821-13230, 33715, 397IB, 33717, 3972B, 39721.
  • the at least one coding sequence of the RNA according to the invention comprises or consists of a nucleic acid sequence identical to or having a sequence identity of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 86%. 87%. 88%, 83%, 30%, 31%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%, preferably of at least 70%.
  • the at least one coding sequence of the RNA according to the invention comprises or consists of a nucleic acid sequence having a sequence identity of at least 8B% with any one of the (modified) nucleic acid sequences defined in SEQ ID NOs: 8821-13230, 39715, 39716, 39717, 3972D, 39721, 39724, 39725, 39728, 39729, 39730, 39733, 39734, 39737, 39738, 39741, 39742, 3B745, 3374B, and/or SEQ 10 NOs: 2B46I-30870, and/or SEQ ID NOs: 30871- 35280, and/or SEQ 10 NOs: 3528I-33B90, and/or SEQ 10 NO: 39713 to SEQ ID NO: 39746, and/or SEQ ID NOs: 39714, 397IB, 39729, 39734, 39738, 39725, or of a fragment or
  • the present invention provides an RNA comprising at least one coding sequence, wherein the coding sequence comprises or consists of a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 39714, 39716, 39729. 39734, 39738, 39725, or a fragment or variant of any one of these nucleic acid sequences.
  • the at least one coding sequence of the RNA according to the invention comprises or consists of a nucleic acid sequence having a sequence identity of at least 5%, 10%.20%, 30%, 40%, 50%. BD%, 70%, 8D%, 85%, 86%.
  • nucleic acid sequence selected from the group consisting of SEQ ID NDs: 39714, 397IB. 39729, 39734, 39738, 39725, or a fragment or variant of any one of these nucleic acid sequences.
  • RNA of the present invention is based on the finding that codons encoding the same amino acid typically occur at different frequencies.
  • the coding sequence (coding region) as defined herein is preferably modified compared to the corresponding region of the respective wild type RNA such that the frequency of the codons encoding the same amino acid corresponds to the naturally occurring frequency of that codon according to the human codon usage as e.g. shown in Table 2.
  • the wild type coding sequence is preferably adapted in a way that the codon "GCC” is used with a frequency of D.4D, the codon “GCT” is used with a frequency of 0.28, the codon “GCA” is used with a frequency of 0.22 and the codon “GCG” is used with a frequency of 0.ID etc. (see Table 2).
  • the present invention provides an RNA comprising at least one coding sequence, wherein the coding sequence comprises a nucleic acid sequence selected from the group consisting of SEQ 10 NOs: 17641-22050, or a fragment or variant of any one of said nucleic acid sequences.
  • the at least one coding sequence of the RNA according to the invention comprises Dr consists of a nucleic acid sequence having a sequence identity of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%, preferably of at least 70%, more preferably of at least 80%, even more preferably at least 85%, even more preferably of at least 9D% and most preferably of at least 95% or even 97%, with a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 17641-22050, or a fragment or variant of any one of said nucleic acid sequences. Codon-optimized sequences:
  • the NA of the present invention comprises at least one coding sequence, wherein the coding sequence is codon-optimized as described herein. More preferably, the codon adaptation index (CAI) of the at least one coding sequence is at least D.5, at least 0.8, at least 0.9 or at least 0.35. Most preferably, the codon adaptation index (CAI) of the at least one coding sequence is I.
  • the wild type coding seqoence is adapted in a way that the most frequent human codon "GCC” is always used for said amino acid, or for the amino acid Cysteine (Cys), the wild type sequence is adapted in a way that the most frequent human cndnn "TGC" is always used for said amino acid etc.
  • the present invention provides an RNA comprising at least one coding sequence, wherein the coding sequence comprises a nucleic acid sequence selected from the group consisting nf SEQ ID NOs: 2205I-2G4B0, D a fragment or variant nf any one of said nucleic acid sequences.
  • the at least one coding sequence of the RNA according to the invention comprises or consists of a nucleic acid sequence having a sequence identity of at least 5%, 10%, 20%, 30%, 40%, 50%, B0%, 70%, 80%, 85%, 86%.
  • the RNA of the composition Df the present invention may be modified by modifying, preferably increasing, the cytosine (C) content of the RNA, preferably of the coding region of the RNA.
  • the C content of the coding region of the RNA of the present invention is modified, preferably increased, compared to the C content of the coding region of the respective wild type RNA, i.e. the unmodified RNA.
  • the amino acid sequence encoded by the at least one coding sequence of the RNA of the present invention is preferably not modified as compared to the amino acid sequence encoded by the respective wild type mRNA.
  • the modified RNA is modified such that at least 10%, 20%, 30%, 40%, 50%, 00%, 70% or 80%. or at least 90% of the theoretically possible maximum cytosine-content or even a maximum cytosine-cDntent is achieved.
  • At least 10%, 20%, 30%, 40%, 50%, 60%. 70%, 80%, 90% or even 100% of the codons of the target RNA wild type sequence, which are "cytosine content optimizable" are replaced by codons having a higher cytosine- content than the ones present in the wild type sequence.
  • some of the codons of the wild type coding sequence may additionally be modified such that a codnn for a relatively rare tRNA in the cell is exchanged by a codon for a relatively frequent tRNA in the cell, provided that the substituted codon for a relatively frequent tRNA carries the same amino acid as the relatively rare tRNA of the original wild type codon.
  • codons for a relatively rare tRNA are replaced by a codon for a relatively frequent tRNA in the cell, except codons encoding amino acids, which are exclusively encoded by codons not containing any cytosine, or except for glutamine (Gin), which is encoded by two codons each containing the same number of cytosines.
  • the modified target RNA is modified such that at least 80%, or at least 90% of the theoretically passible maximum cytosine-content or even a maximum cytosine-content is achieved by means of codons, which code for relatively frequent tRNAs in the cell, wherein the amino acid sequence remains unchanged.
  • more than one codon may encode a particular amioo acid. Accordingly, 18 out of 20 naturally occurring amino acids are encoded by more than one codnn (with Tryp and Met being an exception), e.g. by 2 codons (e.g. Cys, Asp, Glu), by three codons (e.g. lie), by 4 codons (e.g. Al, Gly, Pro) or by G codons (e.g. Leu, Arg, Ser).
  • 2 codons e.g. Cys, Asp, Glu
  • 3 codons e.g. lie
  • 4 codons e.g. Al, Gly, Pro
  • G codons e.g. Leu, Arg, Ser
  • cytosine content-optimizable codon refers to codons, which exhibit a lower content of cytosines than other codons encoding the same amino acid. Accordingly, any wild type codon, which may be replaced by another codon encoding the same amino acid and exhibiting a higher number of cytosines within that codon, is considered to be cytosine-optimizable (C-optimizable). Any such substitution of a C-optimizable wild type codon by the specific C-optimized codon within a wild type coding region increases its overall C content and reflects a C-enriched modified mRNA sequence.
  • the RNA of the present invention preferably the at least one coding sequence of the RNA of the present invention comprises or consists of a C-maximized RNA sequence containing C-optimized codons for all potentially C-optimizable codons. Accordingly, I0D% or all of the theoretically replaceable C-optimizable codons are preferably replaced by C-optimized codons over the entire length of the coding region.
  • cytosine-content optimizable codons are codons, which contain a lower nomber of cytosines than other codons coding for the same amino acid.
  • any of the codons GCG, GCA, GCU codes for the amino acid Ala, which may be exchanged by the codon GCC encoding the same amino acid, and/ or the codon UGU that codes for Cys may be exchanged by the codon UGC encoding the same amino acid, and/or the codon GAU which codes for Asp may be exchanged by the codon GAC encoding the same amino
  • the number of cytosines is increased by I per exchanged codon.
  • Exchange of all non C-optimized codons (corresponding to C-optimizable codons) of the coding region results in a C-maximized coding sequence.
  • at least 70%, preferably at least 80%, more preferably at least 90%, of the non C-optimized codons within the at least one coding region of the NA according to the invention are replaced by C-optimized codons.
  • the percentage of C-optimizable codons replaced by C-optimized codons is less than 70%, while for other amino acids the percentage of replaced codons is higher than 70% to meet the overall percentage of5 C-optimization of at least 70% of all C-optimizable wild type codons of the coding region.
  • a C-optimized RNA of the invention at least 50% of the C-optimizable wild type codons for any given amino acid are replaced by C-optimized codons, e.g. any modified C-enriched RNA preferably contains at least 50% C-optimized codons at C-optimizable wild type codon positions encoding any one of the above mentioned amino acids Ala, Cys, Asp, Phe, Gly, His, He, Leu,0 Asn, Pro, Arg, Ser, Thr, Val and Tyr, preferably at least 60%.
  • codons encoding amino acids which are not cytosine content-optimizable and which are, however, encoded by at least two codons, may be used without any further selection process.
  • the codon of the wild type sequence that codes for a relatively rare tRNA in the cell e.g. a human cell
  • the relatively rare codon GAA coding for Glu may be exchanged by the relative frequent codon GAG coding for the same amino acid
  • the relatively rare codon AAA coding for Lys may be exchanged by the relative frequent codon AAG coding for the same amino acid, and/or
  • the relatively rare codon CAA coding for Gin may be exchanged for the relative frequent codon CAG encoding the same amino acid.
  • the single substitutions listed above may bE used individually as well as in all possible combinations in order to optimize the cytosine-content of the modified RNA compared to the wild type mRNA sequence.
  • the at least one coding sequence as defined herein may be changed compared to the coding region of the respective wild type RNA in such a way that an amino acid encoded by at least two or more codons, of which one comprises one additional cytosine, such a codon may be exchanged by the C-optimized codon comprising one additional cytosine.
  • the amino acid is preferably unaltered compared to the wild type sequence.
  • the present invention provides an RNA comprising at least one coding sequence, wherein the coding sequence comprises a nucleic acid sequence selected from the group consisting of SEQ ID NDs: 13231-17640, or a fragment or variant of any one of said nucleic acid sequences.
  • the at least one coding sequence of the RNA according to the invention comprises or consists of a nucleic acid sequence having a sequence identity of at least 5%, 10%, 20%.30%, 40%, 50%, 60%.70%, 80%, 85%, 8G%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%, preferably of at least 70%, more preferably of at least 8D%, even more preferably at least 85%, even more preferably of at least 90% and most preferably of at least 95% or even 97%, with a nucleic acid sequence selected from the group consisting of SEQ ID NDs: 13231-17640, or a fragment or variant of any one of said nucleic acid sequences.
  • the invention provides an RNA, preferably an mRNA, comprising at least one coding sequence as defined herein, wherein the G/C content of the at least one coding sequence of the RNA is increased compared to the G/C content of the corresponding coding sequence of the corresponding wild type RNA, and/or wherein the C content of the at least one coding sequence of the RNA is increased compared to the G content of the corresponding coding sequence of the corresponding wild type RNA.
  • codons in the at least one coding sequence of the RNA are adapted to human codon usage, wherein the codon adaptation index (CAI) is preferably increased or maximised in the at least one coding sequence of the RNA, and wherein the amino acid sequence encoded by the RNA is preferably not being modified compared to the amino acid sequence encoded by the corresponding wild type RNA.
  • CAI codon adaptation index
  • the artificial nucleic acid, preferably an mRNA, as defined herein, 5 can be modified by the addition of a so-called “5'-cap” structure, which preferably stabilizes the nucleic acid, preferably an mRNA, as described herein.
  • the artificial nucleic acid according to the invention preferably an mRNA, comprises a 5'-cap structure.
  • a 5'-cap is an entity, typically a modified nucleotide entity, which generally "caps" the 5'-end of a nucleic acid, for example of a mature mRNA.
  • a 5'-cap may typically be formed by a modified nucleotide, particularly by a derivative of a guanine nucleotide.
  • the 5'-cap is linked to the 5'-terminus via a 5'-5'-triphosphate linkage.
  • a 5'-cap may be methylated, e.g. m7GpppN, wherein N is the terminal 5' nucleotide of the nucleic acid carrying the 5'-cap, typically the S'-end of an mRNA.
  • m7GpppN is the I5 5'-cap structure, which naturally occurs in mRNA transcribed by polymerase II and is therefore preferably not considered as modification comprised in an artificial nucleic acid in this context.
  • a modified artificial nucleic acid, preferably an mRNA, of the present invention may comprise an m7GpppN as 5'-cap, but additionally the modified artificial nucleic acid, preferably an mRNA, typically comprises at least one further modification as defined herein.
  • 5'cap structures include glyceryl, inverted deaxy abasic residue (moiety), 4',5' methylene nucleotide, I- (beta-D-erythrofuranosyl) nucleotide, 4'-thio nucleotide, carbocyclic nucleotide, l,5-anhydrohexitol nucleotide, L-nucleatides, alpha-nucleotide, modified base nucleotide, threo-pentofuranosyl nucleotide, acyclic 3',4'-seco nucleotide, acyclic 3,4- dihydroxybutyl nucleotide, acyclic 3,5 dihydroxypentyl nucleotide.3'-3'-inverted nucleotide moiety, 3'-3'-inverted abasic moiety, 3'-2'-invertBd nucleotide moiety, 3'-2'
  • modified 5'-cap structures are capl (methylation of the ribose of the adjacent nucleotide of m7G), cap2 (additional methylation of the ribose of the 2nd nucleotide downstream of the m7G), cap3 (additional methylation of the ribose ofD the 3rd nucleotide downstream of the m7G), cap4 (additional methylation of the ribose of the 4th nucleotide downstream of the m7G), ARCA (anti-reverse cap analogue, modified ARCA (e.g.
  • phosphothioate modified ARCA inosine, Nl-methyl-guanosine, 2'- fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine. LNA-guanosine. and 2-azido-guanosine.
  • the artificial nucleic acid comprises an untranslated region (UTR). More preferably, the artificial nucleic acid according to the invention, preferably an mRNA. comprises at least one of the following structural elements: a 5'- and/or 3'- untranslated region element (UTR element), particularly a 5'-UTR element, which comprises or consists of a nucleic acid sequence which is derived from the 5'-UTR of a TOP gene or from a fragment, homolog or a variant thereof, or a 5'- and/ or 3'-UTR element which may be derivable from a gene that provides a stable mRNA or from a homolog, fragment or variant thereof; a histone-stem-loop structure, preferably a histone-stem-loop in its 3' untranslated region; a 5'-cap structure; a poly- 5 A tail; or a poly(C) sequence.
  • UTR element 5'- and/or 3'- untranslated region element
  • a 5'-UTR element
  • the artificial nucleic acid preferably an mRNA, comprises at least one 5'- or 3'-UTR element.
  • an UTR element comprises or consists of a nucleic acid sequence, which is derived from the 5'- or 3'-UTR of any naturally occurring gene or which is derived from a fragment, a homolog or a variant of the 5'- or 3'-UTR of a gene.
  • the 5'- or ID 3'-UTR element used according to the present invention is heterologous to the coding region of the inventive artificial nucleic acid. Even if 5'- or 3'-UTR elements derived from naturally occurring genes are preferred, also synthetically engineered UTR elements may be used in the context of the present invention.
  • the artificial nucleic acid according to the invention comprises a 5'-UTR. More preferably, 15 the artificial nucleic acid comprises a 5'-UTR comprising at least one heterologous 5'-UTR element.
  • the artificial nucleic acid comprises at least one 5'-untranslated region element (5'-UTR element), preferably a heterologous 5'-UTR element, which comprises or consists of a nucleic acid sequence, which is derived from the 5'-UTR of a TOP gene or which is derived from a fragment, homolog or variant of the 5'-UTR of a TOP gene.
  • 5'-UTR element preferably a heterologous 5'-UTR element, which comprises or consists of a nucleic acid sequence, which is derived from the 5'-UTR of a TOP gene or which is derived from a fragment, homolog or variant of the 5'-UTR of a TOP gene.
  • the 5'-UTR element does not comprise a TDP-motif Dr a 5'TQP, as defined above.
  • the nucleic acid sequence of the 5'-UTR element which is derived from a 5'-UTR of a TDP gene, terminates at its 3'-end with a nucleotide located at position I, Z, 3, 4, 5, G, 7, 8, 9 or ID upstream of the start codon (e.g. A(U/T)G) of the 25 gene or mRNA it is derived from.
  • the 5'-UTR element does not comprise any part of the protein coding region.
  • the only protein coding part of the artificial nucleic acid is provided by the at least one coding region.
  • the nucleic acid sequence which is derived from the 5'-UTR of a TDP gene, is typically derived from a eukaryotic TDP gene, preferably a plant or animal TDP gene, more preferably a chardate TDP gene, even more preferably a vertebrate TDP gene, most 3D preferably a mammalian TDP gene, such as a human TDP gene.
  • the 5'-UTR element is preferably selected from 5'-UTR elements comprising or consisting of a nucleic acid sequence, which is derived from a nucleic acid sequence selected from the group consisting of SED ID NDs: I-I3B3, SED ID NO: 1305, SED ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO 2013/143700, whose disclosure is incorporated herein by reference, 35 from the homologs of SEQ ID NDs: 1-1363, SEQ ID NO: 1395, SEO ID ND: 1421 and SEQ ID NO: 1422 of the patent application WO 2013/143700, from a variant thereof, or preferably from a corresponding RNA sequence.
  • the 5'-UTR element of the artificial nucleic acid preferably an mRNA, comprises or consists of a nucleic acid sequence, which is derived from a nucleic acid sequence extending from nucleotide position 5 (i.e.
  • the 5'-0TR element is derived from a nucleic acid sequence extending from the nucleotide position immediately 3' to the 5T0P to the nucleotide position immediately 5' to the start codon (located at the 3'-end of the sequences), e.g.
  • nucleotide position immediately 5' to the ATG sequence of a nucleic acid sequence selected from SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO 2013/143700, from the homologs of SEQ ID NOs: 1-1363, SEQ ID NO: 1395, SEQ ID NO: 1421 and SEQ ID NO: 1422 of the patent application WO 2013/143700, from a variant thereof, or a corresponding RNA sequence.
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from a 5'-UTR of a TOP gene encoding a ribosomal protein or from a variant of a 5'-UTR of a TOP gene encoding a ribosomal protein.
  • the 5'-0TR element comprises or consists of a nucleic acid sequence, which is derived from a 5'-UTR of a nucleic acid sequence according to any of SEQ ID NO: 67, 170, 193, 244, 259, 554, 650, 675, 700, 721, 913, 1016, 1063, 1120, 1138.
  • sequences of the invention are selected from the group of any of SEQ 10 NOs: 1-4410, or a fragment or variant of any of these sequences, wherein these sequences resemble VPI protein sequences.
  • sequences of the invention are selected from the group of any of SEQ ID NOs: 4411-8820, 39713, 30714, 3B7I8, 39719, 39722, 39723, 39726, 39727, 39731, 39732, 39735, 39736, 39739, 39740, 39743 and 39744, or a fragment or variant of any of these sequences, wherein these sequences resemble VPI nucleotide wild type sequences.
  • sequences of the invention are selected from the group of any of SEQ ID NOs: 8821-13230, 39715, 3B7I6, 33717, 39720, 33721, 33724, 33725. 39728, 33723, 33730, 39733, 39734, 39737, 39738, 39741, 39742, 39745, 3974G.
  • sequences of the invention are selected from the group of any of SEQ ID NDs: I323I-I7B4D, or a fragment 5 or variant of any of these sequences, wherein these sequences resemble optimized VPI nucleotide wild type sequences.
  • sequences of the invention are selected from the group of any of SEQ ID NDs: I7B4I-22050. or a fragment or variant of any of these sequences, wherein these sequences resemble optimized VPI nucleotide wild type sequences.
  • sequences of the invention are selected from the group of any of SEQ ID NDs: 22D5I-2B4BD, or a fragment or variant of any of these sequences, wherein these sequences resemble optimized VPI nucleotide wild type sequences.
  • sequences of the invention are selected from the group of any of SED ID NDs: 2B4BI-3D87D, or a fragment or variant of any of these sequences, wherein these sequences resemble optimized VPI nucleotide wild type sequences.
  • sequences of the invention are selected from the group of any of SEQ ID NDs: 3D87I-3528Q, or a fragment or variant of any of these sequences, wherein these sequences resemble optimized VPI nucleotide wild type sequences.
  • sequences of the invention are selected from the group of any of SEQ ID NDs: 35281-39690, or a fragmentD or variant of any of these sequences, wherein these sequences resemble optimized VPI nucleotide wild type sequences.
  • the artificial nucleic acid according to the invention comprises a 5'-UTR comprising at least one heterologous 5'-DTR sequence, wherein the at least one heterologous 5'-UTR element comprises a nucleic acid sequence, which is derived from a 5'- UTR of a TOP gene encoding a ribosomal protein, preferably from a corresponding RNA sequence, or from a homolog, a fragment5 or a variant thereof, preferably lacking the 5'TOP motif.
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from a 5'-DTR of a TOP gene encoding a ribosomal Large protein (RPL) or from a homolog Dr variant of a 5'-UTR of a TDP gene encoding a ribosomal Large protein (RPL).
  • RPL ribosomal Large protein
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from a 5'-DTRD of a nucleic acid sequence according to any of SEQ ID NDs: 67, 259, 1284-1318, 1344, 1346, 1348-1354, 1357, 1358, 1421 and 1422 of the patent application WD 2013/143700, a corresponding RNA sequence, a homolog thereof, or a variant thereof as described herein, preferably lacking the 5'TOP motif.
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived5 from the 5'-0TR of a ribosomal protein Large 32 gene, preferably from a vertebrate ribosomal protein Large 32 (L32) gene, more preferably from a mammalian ribosomal protein Large 32 (L32) gene, most preferably from a human ribosomal protein Large 32 (L32) gene, or from a variant of the 5'-UTR of a ribosomal protein Large 32 gene, preferably from a vertebrate ribasomal protein Large 32 (L32) gene, more preferably from a mammalian ribosomal protein Large 32 (L32) gene, most preferably from a human ribosomal protein Large 32 (L32) gene, wherein preferably the 5'-UTR element does not comprise the 5'TOP of said gene.
  • the 5'-UT element comprises or consists of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 00%, even more preferably of at least about 95%, even more preferably of at least about 03% to the nucleic acid sequence according to SEO ID NO: 20649 (5'-UTR of human ribosomal protein Large 32 lacking the 5'-terminal oligopyrimidine tract; corresponding to SEO ID NO: 1368 of the patent application W02013/143700) or preferably to a corresponding RNA sequence, such as SEO ID NO: 39692, or wherein the at least one 5'-UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 7D%
  • the fragment exhibits a length of at least about 20 nucleotides or more, preferably of at least about 30 nucleotides or more, more preferably of at least about 40 nucleotides or more.
  • the fragment is a functional fragment as described herein.
  • the artificial nucleic acid according to the invention comprises a 5'-0TR element, which comprises or consists of a nucleic acid sequence, which is derived from the 5'-0TR of a vertebrate TOP gene, such as a mammalian, e.g. a human TOP gene, selected from RPSA, RPS2, RPS3, RPS3A, RPS4, RPS5.
  • a mammalian e.g. a human TOP gene
  • RPS6 RPS7, RPS8. RPS9, RPSI0, RPSII, RPSI2, RPSI3, RPSI4, RPSI5, RPSI5A, RPSI6, RPSI7, RPSI8, RPSI9, RPS20, RPS2I.
  • RPLP0 RPLPI, RPLP2, RPLP3, RPLP0, RPLPI, RPLP2, EEFIAI, EEFIB2, EEFID, EEFIG, EEF2, EIF3E, EIF3F, EIF3H, EIF2S3, EIF3C, EIF3K, EIF3EIP, EIF4A2, PABPCI, HNRNPAI, TPTI, TUBBI, 0BA52, NP I.
  • the 5'-0TR element does not comprise a TDP-motif or the 5'TOP of said genes, and wherein optionally the 5'-UTR element starts at its 5'-end with a nucleotide located at position 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 downstream of the 5'-terminal oligopyrimidine tract (TOP) and wherein further optionally the 5'-UTR element which is derived from a 5'-0TR of a TOP gene terminates at its 3'-end with a nucleotide located at position I, 2, 3, 4, 5, 6, 7, 8, 9 or ID upstream of the start codon (A(U/T)G) of the gene it is derived from.
  • TOP 5'-terminal oligopyrimidine tract
  • the artificial nucleic acid comprises at least one heterologous 5'-UTR element comprising a nucleic acid sequence, which is derived from a 5'-0TR of a TDP gene encoding a ribosomal Large protein (RPL), preferably PL32 or RPL35A, ar from a gene selected from the group consisting of HSDI7B4, ATP5AI, AIGI. ASAHI, CDXBC or ABCB7 (also referred to herein as MDR), or from a homolog, a fragment or variant of any one of these genes, preferably lacking the 5'TDP motif.
  • RPL ribosomal Large protein
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, which is derived from the 5'-UTR of a ribosomal protein Large 32 gene (RPL32).
  • RPL35 a ribosomal protein Large 35 gene
  • RPL2I a ribosomal protein Large 21 gene
  • AIGI cytochrome c oxidase subunit Vic gene
  • CDXBC cytochrome c oxidase subunit Vic gene
  • ASAHI N-acylsphingosine amidohydrolase
  • RPL32 vertebrate ribosomal protein Large 32 gene
  • RPL35 vertebrate ribosomal protein Large 35 gene
  • RPL2I vertebrate ribosomal protein Large 21 gene
  • AIGI vertebrate cytochrome c oxidase subunit Vic gene
  • CDXBC vertebrate N-acylsphingosine amidohydrolase
  • ASAHI vertebrate ATP-Binding Cassette, Sub-Family B (MDR/TAP), Member 7 gene (ABCB7), or from a variant thereof, more preferably from a mammalian ribosomal protein Large 32 gene (RPL32), a ribosomal protein Large 35 gene (RPL35), a ribosomal protein Large 21 gene (RPL2I), a mammalian ATP synthase, H+ transporting, mitochondrial Fl complex, alpha subunit I, cardiac muscle (ATP5AI) gene, a mammalian hydroxysteroid (17-beta) dehydrogenase 4 gene (HSDI7B4), a mammalian androgen-induced I gene (AIGI), a mammalian cyto-chrome c oxidase subunit Vic gene (CDXBC), a mammalian N-acylsphingosine0 ami-dohydrolase (acid cerami)
  • H+ transporting mitochondrial Fl complex, alpha subunit I, cardiac muscle (ATP5AI) gene, a human hydroxysteroid (17-beta) dehydrogenase 4 gene (HSDI784), a human androgen-induced I gene (AIGI), a human cytochrome c oxidase subunit Vic gene5 (CDXBC), a human N-acylsphingosine amidohydrolase (acid ceramidase) I gene (ASAHI), or a human ATP-Binding Cassette, Sub- Family B (MDR/TAP).
  • Member 7 gene (ABCB7), or from a variant thereof, wherein preferably the 5'-DTR element does not comprise the 5'TDP of said gene.
  • the 5'-UTR element comprises or consists of a nucleic acid sequence, whichD has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 30%, even more preferably of at least about 95%, even more preferably of at least about 93% to the nucleic acid sequence according to SED ID NO: I3G8, or SED ID NOs: 1412-1420 of the patent application WD 2013/143700, or a corresponding RNA sequence, or wherein the at least one 5'-UTR element comprises or consists of a fragment of a nucleic acid sequence which has an identity of at least about 40%, preferably5 of at least about 50%, preferably of at least about 60%.
  • the fragment is as described above, i.e. being a continuous stretch of nucleotides representing at least 20% etc. of the full-length 5'-UTR.
  • the fragment exhibits a length of at least about 2D nucleotides or more, preferably of at least about 3D nucleotides or more, more preferably of at least about 40 nucleotides or more.
  • the fragment is a functional fragment as described herein.
  • the artificial nucleic acid comprises a 5'-UTR comprising at least one heterologous 5'-0TR element, wherein the heterologous 5'-0TR element comprises a nucleic acid sequence according to SEO ID ND: 39G9I to SEO ID ND: 39FJ94. or a homolog, a fragment or a variant thereof.
  • the at least one heterologous 5'-UTR element comprises or consists of a nucleic acid sequence, which has an identity of at least about 40%, preferably of at least about 50%, preferably of at least about 60%, preferably of at least about 70%, more preferably of at least about 80%, more preferably of at least about 90%, even more preferably of at least about 95%, even more preferably of at least about 99% to a nucleic acid sequence according to any one of SEQ ID NO: 33631 to SEQ ID ND: 39694.
  • the artificial nucleic acid according to the invention comprises a 3'-untranslated region (S'-UTR). More preferably, the artificial nucleic acid according to the invention comprises a 3'-0TR comprising or consisting of at least one heterologous 3'-0TR element, preferably as defined herein.
  • the artificial nucleic acid may contain a poly-A tail of typically about ID to 200 adenosine nucleotides, preferably about ID to 100 adenosine nucleotides, more preferably about 40 to 8D adenosine nucleotides or even more preferably about 50 to 7D adenosine nucleotides.
  • the poly(A) sequence in the artificial nucleic acid is derived from a DNA template by in vitro transcription.
  • the poly(A) sequence may also be obtained in vitro by common methods of chemical-synthesis without being necessarily transcribed from a DNA progenitor.
  • poly(A) sequences, or poly(A) tails may be generated by enzymatic polyadenylation of the RNA according to the present invention using commercially available polyadenylation kits and corresponding protocols known in the art, or using immobilized poly(A)polymerases e.g. in a polyadenylation reactor (WD 2016/174271).
  • the artificial nucleic acid optionally comprises a polyadenylation signal, which is defined herein as a signal, which conveys polyadenylation to a (transcribed) mRNA by specific protein factors (e.g. cleavage and polyadenylation specificity factor (CPSF), cleavage stimulation factor (CstF), cleavage factors I and II (CF I and CF II), poly(A) polymerase (PAP)).
  • CPSF cleavage and polyadenylation specificity factor
  • CstF cleavage stimulation factor
  • CF I and CF II cleavage factors I and II
  • PAP poly(A) polymerase
  • a consensus polyadenylation signal is preferred comprising the NN(U/T)ANA consensus sequence.
  • the polyadenylation signal comprises one of the following sequences: AA(U/T)AAA or A(0/T)(U/T)AAA (wherein uridine is usually present in RNA and thymidine is usually present in DNA).
  • the artificial nucleic acid of the present invention preferably the S'-UTR of the artificial nucleic acid, may contain a poly-C tail of typically about ID to 20D cytosine nucleotides, preferably about ID to IDO cytosine nucleotides, more preferably about 20 to 70 cytosine nucleotides or even more preferably about 20 to GO or even 10 to 40 cytosine nucleotides.
  • the artificial nucleic acid according to the invention further comprises at least one S'-UTR element, which comprises or consists of a nucleic acid sequence derived from the 3'-UTR of a chordate gene, preferably a vertebrate genE, more preferably a mammalian gene, most preferably a human gene, or from a variant of the 3'-UT of a chordate gene, preferably a vertebrate gene, more preferably a mammalian gene, most preferably a human gene.
  • S'-UTR element which comprises or consists of a nucleic acid sequence derived from the 3'-UTR of a chordate gene, preferably a vertebrate genE, more preferably a mammalian gene, most preferably a human gene, or from a variant of the 3'-UT of a chordate gene, preferably a vertebrate gene, more preferably a mammalian gene, most preferably a human gene.
  • the tfirm "3'-0TR element” refers to a nucleic acid sequence, which comprises or consists of a nucleic acid sequence that is derived from a 3'-UTR or from a variant of a S'-UTR.
  • a 3'-DTR element in the sense of the present invention may represent the 3'-DTR on a DNA or on an RNA level.
  • a 3'-DTR element may be the 3'-UTR of an mRNA, preferably nf an artificial mRNA, or it may be the transcription template for a 3'-UTR of an mRNA.
  • a 3'-UTR element preferably is a nucleic acid sequence, which corresponds to the 3'-UTR of an mRNA, preferably to the S'-UTR of an artificial mRNA, such as an mRNA obtained by transcription of a genetically engineered vector construct.
  • the 3'-UTR element fulfils the function of a 3'-UTR or encodes a sequence, which fulfils the function of a 3'-DTR.
  • the artificial nucleic acid comprises a 3'-UTR element comprising or consisting of a nucleic acid sequence derived from a S'-UTR of a gene, which preferably encodes a stable mRNA, or from a homolog, a fragment Dr a variant of said gene.
  • the 3'-UTR element may be derivable from a gene that relates to an mRNA with an enhanced half-life (that provides a stable mRNA), for example a 3'-UTR element as defined and described below.
  • the 3'-UTR element comprises or consists of a nucleic acid sequence which is derived from a 3'-UTR of a gene selected from the group consisting of an albumin gene, an a-globin gene, a ⁇ -globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, such as a collagen alpha 1(1) gene, or from a homolog, a fragment or a variant of a 3'-DTR of a gene selected from the group consisting of an albumin gene, an a-globin gene, a ⁇ -globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, such as a collagen alpha 1(1) gene.
  • the 3'-DTR element comprises or consists of a nucleic acid sequence which is derived from a 3'-UTR of a gene selected from the group consisting of an albumin gene, an a-globin gene, a ⁇ -globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, such as a collagen alpha 1(1) gene, or from a homolog, a fragment or a variant of a 3'-UTR of a gene selected from the group consisting of an albumin gene, an ⁇ -globin gene, a ⁇ -globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, and a collagen alpha gene, such as a collagen alpha 1(1) gene according to SE0 ID NOs: I3G9-I300 of the patent application WD 2013/143700, whose disclosure is incorporated herein by reference, or from a homalag, a fragment or
  • the 3'-0TR element comprises or consists of a nucleic acid sequence, which is derived from the 3'-UTR of a vertebrate albumin gene or from a variant thereof, preferably from the 3'-0TR of a mammalian albumin gene or from a variant thereof, more preferably from the S'-UT of a human albumin gene or from a variant thereof, even more preferably from the 3'-0TR of the human albumin gene according to Genbank Accession number NM_000477.5, or from a fragment or variant thereof.
  • the 3'-UTR element comprises or consists of a nucleic acid according to SEO ID NO: 39703, or SEO ID NO: 39704 (corresponding to SEQ ID NO: I3B9 of the patent application WO 2013/143700), or a fragment, homolog or variant thereof.
  • the 3'-0TR element comprises or consists of the nucleic acid sequence derived from a fragment of the human albumin gene according to SEQ ID NO: 39705, or SEO ID NO: 39706 (corresponding to SEQ ID NO: I37G of the patent application W0 2013/143700), or a fragment, homolog or variant thereof.
  • the 3'-0TR element comprises or consists of a nucleic acid according to SEO ID NO: 39707, or SEQ ID NO: 397D8, (Albumin 7), or a fragment, homolog or variant thereof.
  • the at least one heterologous 3'-0TR element comprises or consists of a nucleic acid sequence derived from a 3'-UTR of an a-globin gene, preferably a vertebrate a-or ⁇ -globin gene, more preferably a mammalian a-or ⁇ -globin gene, most preferably a human a-or ⁇ -globin gene.
  • the 3'-0TR element comprises or consists of a nucleic acid according to SEQ ID NO: 39G95, or SEQ ID NO: 39G9B (corresponding to SEQ ID NO: 1370 of the patent application W0 2013/143700). or a homolog, a fragment, or a variant thereof
  • the at least one heterologous 3'-0TR element comprises or consists of a nucleic acid sequence derived from a 3'- 0TR of Homo sapiens hemoglobin, alpha I (HBAI). More preferably, the 3'-0TR element comprises or consists of a nucleic acid according to SEQ ID ND: 39695, or SEQ ID NO: 3969G (corresponding to SEQ ID NO: I37D of the patent application W0 2013/143700), or a homolog, a fragment, or a variant thereof.
  • the at least one heterologous 3'-UTR element comprises or consists of a nucleic acid sequence derived from a 3'-UTR of Homo sapiens hemoglobin, alpha 2 (HBA2). More preferably, the 3'-UTR element comprises or consists of a nucleic acid according to SEQ ID NO: 39697 or SEO ID NO: 39698 (corresponding to SEQ ID NO: 1371 of the patent application W0 20I3/I4370D), or a homolog, a fragment, or a variant thereof.
  • the at least one heterologous 3'-0TR element comprises or consists of a nucleic acid sequence derived from a 3'-0TR of Homo sapiens hemoglobin, beta (HBB). More preferably, the 3'-0TR element comprises or consists of a nucleic acid according to SEQ ID NO: 39609, or SED ID NO: 39700 (corresponding to SEO ID NO: 1372 of the patent application WO 2013/143700), or a homolog, a fragment, or a variant thereof.
  • the at least one heterologous 3'-DTR element may further comprise or consist of the center, ⁇ -complex-binding portion of the 3'-0TR of an a-globin gene, such as of a human a-globin gene, or a homolog, a fragment, or a variant of an a-globin gene, preferably according to SEQ ID NO: 39701 or SEO ID NO: 39702 (also referred to herein as "muag”) (corresponding to SEO ID NO: 1393 of the patent application WO 2013/143700), or a homolog, a fragment, or a variant thereof.
  • an a-globin gene such as of a human a-globin gene, or a homolog, a fragment, or a variant of an a-globin gene, preferably according to SEQ ID NO: 39701 or SEO ID NO: 39702 (also referred to herein as "muag") (corresponding to SEO ID NO: 1393 of the patent application WO 2013/143700), or
  • a nucleic acid sequence which is derived from the 3'-0TR of a preferably refers to a nucleic acid sequence which is based on the 3'-UTR sequence of a noted gene or on a part thereof, such as on the 3'-UTR of an albumin gene, an a-globin gene, a ⁇ -globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, or a collagen alpha gene, such as a collagen alpha 1(1) gene, preferably of an albumin gene or on a part thereof.
  • This term includes sequences corresponding to the eotire 3'-0TR sequence, i.e.
  • the full length 3'-0TR sequence of a gene and sequences corresponding to a fragment of the 3'-UTR sequence of a gene, such as an albumin gene, ⁇ -globin gene, ⁇ -globin gene, tyrosine hydroxylase gene, lipoxygenase gene, or collagen alpha gene, such as a collagen alpha 1(1) gene, preferably of an albumin gene.
  • a gene such as an albumin gene, ⁇ -globin gene, ⁇ -globin gene, tyrosine hydroxylase gene, lipoxygenase gene, or collagen alpha gene, such as a collagen alpha 1(1) gene, preferably of an albumin gene.
  • a nucleic acid sequence which is derived from a variant of the 3'-UTR of a pot gene preferably refers to a nucleic acid sequence, which is based on a variant of the 3'-0TR sequence of a gene, such as on a variant of the 3'-UTR of an albumin gene, an a-globin gene, a ⁇ -globin gene, a tyrosine hydroxylase gene, a lipoxygenase gene, or a collagen alpha gene, such as a collagen alpha 1(1) gene, or on a part thereof as described above.
  • This term includes sequences corresponding to the entire sequence of the variant of the 3'-UTR of a gene, i.e.
  • a fragment in this context preferably consists of a continuous stretch of nucleotides corresponding to a continuous stretch of nucleotides in the full-length variant 3'-UTR, which represents at least 20%, preferably at least 30%, more preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, even more preferably at least 70%, even more preferably at least 80%, and most preferably at least 90% of the full-length variant 3'-0TR.
  • Such a fragment of a variant in the sense of the present invention, is preferably a functional fragment of a variant as described herein.
  • the at least one 5'-UTR element and the at least one 3'-0TR element act synergistically to increase protein production from the inventive artificial nucleic acid as described above.
  • the inventive artificial nucleic acid as described herein comprises a histone stem-loop sequence/structure (histone stem-loop).
  • histone stem-loop sequences are preferably selected from histone stem-loop sequences as disclosed in W02012/019780, whose disclosure is incorporated herewith by reference.
  • a histnne stem-loop sequence, suitable to be used within the present invention is preferably selected from at least one of the following formulae (I) or (II): formula (I) (stem-loop sequence without stem bordering elements):
  • steml or stem2 bordering elements NI-B is a consecutive sequence of I to B, preferably of 2 to 6, more preferably of 2 to 5, even more preferably of 3 to 5, most preferably of 4 to 5 or 5 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C, or a nucleotide analogue thereof:
  • steml [N0-2GN3.5] is reverse complementary or partially reverse complementary with element stem2, and is a consecutive sequence between of 5 to 7 nucleotides;
  • No-z is a consecutive sequence of D to 2, preferably of D to I, mare preferably of I N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof:
  • N3-5 is a consecutive sequence of 3 to 5, preferably of 4 to 5, more preferably of 4 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T
  • each No-4 is independent from another a consecutive sequence of D to 4, preferably of I to 3, more preferably of I to 2 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof;
  • U/T represents uridine, or optionally thymidine
  • stem2 [N3-5CN0-2] is reverse complementary or partially reverse complementary with element steml, and is a consecutive sequence between of 5 to 7 nucleotides; wherein N3-5 is a consecutive sequence of 3 to 5.
  • each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof;
  • No-z is a consecutive sequence of 0 to 2, preferably of D to I, mare preferably of 1 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G or C or a nucleotide analogue thereof; and wherein C is cytidine or an analogue thereof, and may be optionally replaced by a guanosine or an analogue thereof provided that its complementary nucleoside guanosine in steml is replaced by cytidine; wherein
  • steml and stem2 are capable of base pairing with each other forming a reverse complementary sequence, wherein base pairing may occur between steml and stem2, e.g. by Watson-Crick base pairing of nucleotides A and U/T or G and C or by non-Watson- Crick base pairing e.g. wobble base pairing, reverse Watson-Crick base pairing, Hoogsteen base pairing, reverse Hoogsteen base pairing or are capable of base pairing with each other forming a partially reverse complementary sequence, wherein an incomplete base pairing may occur between steml and stem2. on the basis that one ore more bases in one stem do not have a complementary base in the reverse complementary sequence of the other stem.
  • the inventive artificial nucleic acid may comprise at least one histone stem-loop sequence according to at least one of the following specific formulae (la) or (Ha): formula (la) (stem-loop sequence without stem bordering elements):
  • bordering element bordering element wherein: N, C, G, T and U are as defined above.
  • the inventive artificial nucleic acid may comprise at least one histone stem-loop sequence according to at least one of the following specific formulae (lb) or (lib): formula (lb) (stem-loop sequence without stem bordering elements):
  • steml loop stem2 formula (lib) (stem-loop sequence with stem bordering elements):
  • bordering element bordering element wherein: N. C, G, T and U are as defined above.
  • a particular preferred histone stem-loop sequence is the nucleic acid sequence according to SEQ ID NO: 30703, or more preferably the corresponding NA sequence according to SEQ ID NO: 3B7I0. Additional peptide or protein elements:
  • the artificial nucleic acid sequence may additionally encode further peptide or protein elements that e.g., promote secretion of the protein (secretory signal peptides), promote anchoring of the encoded antigen in the plasma membrane (transmembrane domains), promote virus- like particle formation (VLP forming domains).
  • the artificial nucleic acid sequence according to the present invention may additionally encode peptide linker elements, self-cleaving peptides or helper peptides.
  • the inventive artificial nucleic acid may additionally or alternatively encode a secretory signal peptide (signal sequence).
  • signal peptides are sequences, which typically exhibit a length of about ID to 3D amino acids and are preferably located at the H-terminus of the encoded peptide, without being limited thereto.
  • Signal peptides as defined herein preferably allow the transport of the at least one protein encoded by the at least one coding region of the inventive artificial nucleic acid into a defined cellular compartment, preferably the cell surface, the endoplasmic reticulum (ER) or the endosomal-lysosomal compartment.
  • secretory signal peptide sequences as defined herein include, without being limited thereto, signal sequences of classical or non-classical MHC-molecules (e.g. signal sequences of HC I and II molecules, e.g. of the MHC class I molecule HLA-A*D2DI), signal sequences of cytokines or immunoglobulines as defined herein, signal sequences of the invariant chain of immunoglobulines or antibodies as defined herein, signal sequences of Lampl, Tapasin, Erp57, Calretikulin, Calnexin, and further membrane associated proteins or of proteins associated with the endoplasmic reticulum (ER) or the endosomal-lysosomal juxtapos. More preferably, signal sequences of MHC class I molecule HLA-A*020I may be used according to the present invention.
  • signal sequences of MHC class I molecule HLA-A*020I may be used according to the present invention.
  • the artificial nucleic acid sequence particularly the RNA sequence according to the invention may additionally encode at least one transmembrane domain element.
  • Transmembrane elements or membrane spanning polypeptide elements are present in proteins that are integrated or anchored in plasma membranes of cells.
  • Typical transmembrane elements are alpha-helical transmembrane elements.
  • Such transmembrane elements are composed essentially of amino acids with hydrophobic side chains, because the interior of a cell membrane (lipid bilayer) is also hydrophobic.
  • transmembrane elements are commonly single hydrophobic alpha helices or beta barrel structures; whereas hydrophobic alpha helices are usually present in proteins that are present in membrane anchored proteins (e.g., seven transmembrane domain receptors), beta-barrel structures are often present in proteins that generate pores or channels.
  • target proteins such as antigenic peptides or proteins according to the present invention (derived from Norovirus) it may be beneficial to introduce a transmembrane element into the respective constructs.
  • a transmembrane element to the target peptide/protein it may be possible to further enhance the immune response, wherein the translated target peptide/protein, e.g. a viral antigen, anchors to a target membrane, e.g. the plasma membrane of a cell, thereby increasing immune responses. This effect is also referred to as antigen clustering.
  • transmembrane element When used in combination with a polypeptide or protein of interest in the context of the present invention, such transmembrane element can be placed N-terminal or C-terminal to the Norovirus antigenic peptide or protein of interest.
  • the coding sequence far such transmembrane element is typically placed in frame (i.e. in the same reading frame), 5' or 3' to the coding sequence of the polypeptide as defined herein.
  • the transmembrane domain may be selected from the transmembrane domain of Hemagglutinin (HA) of Influenza virus, Env of HIV-I, EIAV (equine infectious anaemia virus), MLV (murine leukaemia virus), mouse mammary tumor virus, G protein of VSV (vesicular stomatitis virus). Rabies virus, or a transmembrane element of a seven transmembrane domain receptor.
  • HA Hemagglutinin
  • EIAV equine infectious anaemia virus
  • MLV murine leukaemia virus
  • mouse mammary tumor virus G protein of VSV (vesicular stomatitis virus).
  • Rabies virus or a transmembrane element of a seven transmembrane domain receptor.
  • the artificial nucleic acid sequence particularly the RNA sequence according to the invention may additionally encode at least Dne VLP forming domain.
  • VLPs are self-assembled viral structural proteins (envelope proteins or capsid proteins) that structurally resemble viruses (without containing viral genetic material). VLPs contain repetitive high density displays of antigens which present conformational epitopes that can elicit strong T cell and B cell immune responses.
  • VLP forming element When used in combination with a Norovirus antigenic peptide or protein in the context of the present invention, such VLP forming element can be placed N-terminal or C-terminal to the polypeptide nf interest.
  • the coding sequence for such VLP forming element is typically placed in frame (i.e. in the same reading frame), 5' or 3' to the coding sequence of the polypeptide as defined herein.
  • RNA nucleic acid
  • Norovirus antigenic polypeptides or proteins it may be beneficial to introduce a VLP forming element into the respective constructs.
  • an improved secretion of the VLP particle may also increase the immunogenicity of the respective antigen.
  • VLP forming elements fused to an antigen may generate virus like particles containing repetitive high density displays of antigens.
  • VLP farming elements can be chosen from any viral or phage capsid or envelope protein.
  • the artificial nucleic acid sequence particularly the RNA sequence according to the invention may additionally encode at least one peptide linker element.
  • the protein elements may be separated by peptide linker elements. Such elements may be beneficial because they allow for a proper folding of the individual elements and thereby the proper functionality of each element.
  • the term "spacer” or "peptide spacer” is used herein.
  • linkers or spacers are particularly useful when encoded by a nucleic acid encoding at least two functional protein elements, such as at least one polypeptide or protein of interest (Norovirus antigens) and at least one further protein or polypeptide element (e.g., VLP forming domain, transmembrane domain).
  • the 5 linker is typically located on the polypeptide chain in between the polypeptide of interest and the at least one further protein element.
  • the coding sequence for such linker is typically placed in the reading frame, 5' or 3' to the coding sequence for the polypeptide Dr protein of interest, or placed between coding regions for individual polypeptide domains of a given protein of interest.
  • ID Peptide linkers are preferably composed of small, non-polar (e.g. Gly) or polar (e.g. Ser or Thr) amino acids. ThB small size of these amino acids provides flexibility, and allows for mobility of the connecting functional domains. The incorporation of Ser or Thr can maintain the stability of the linker in aqueous solutions by farming hydrogen bonds with the water molecules, and therefore reduces an interaction between the linker and the protein moieties. Rigid linkers generally maintain the distance between the protein domains and they may be based on helical structures and/or they have a sequence that is rich in proline.
  • Cleavable linkers allow for in vivo separation of the protein domains.
  • the mechanism of cleavage may be based e.g. on reduction of disulfide bonds within the linker sequence or proteolytic cleavage.
  • the cleavage may be mediated by an enzyme (enzymatic cleavage), e.g. the cleavage linker may provide a protease sensitive sequence (e.g., furin cleavage).
  • D A typical sequence of a flexible linker is composed of repeats of the amino acids Glycine (G) and Serine (S).
  • the linker may have the following sequence: GS, GSG, SGG, SG, GGS, SGS, GSS, SSG. In some embodiments, the same sequence is repeated multiple times (e.g. two, three, four, five or six times) to create a longer linker. In other embodiments, a single amino acid residue such as S or G can be used as a linker. 5 Linkers or spacers may be used as additional elements to promote or improve the secretion of the target protein (Norovirus antigenic peptides or proteins).
  • the artificial nucleic acid sequence particularly the RNA sequence according to the invention may additionally encode at least one self-cleaving peptide.
  • Viral self-cleaving peptides allow the expression of multiple proteins from a single open reading frame.
  • the terms 2A peptide and 2A element are used interchangeably herein.
  • the mechanism by the 2A sequence for generating two proteins from one transcript is by ribosome skipping - a normal peptide bond is impaired at 2A, resulting in two discontinuous protein fragments from one translation event.
  • such 2A peptides are particularly useful when encoded by a nucleic acid encoding at least two functional protein elements (e.g. two Norovirus antigenic peptides or proteins).
  • a 2A element is useful when the nucleic acid molecule encodes at least one polypeptide or protein of interest and at least one further protein element.
  • a 2A element is present when the polynucleotide of the invention encodes two proteins or 5 polypeptides of interest, e.g. two antigens.
  • the coding sequence for such 2A peptide is typically located in between the coding sequence of the polypeptide of interest and the coding sequence of the least one further protein element (which may also be a polypeptide of interest), so that cleavage of the 2A peptide leads to two separate polypeptide molecules, at least one of them being a polypeptide or protein of interest.
  • target proteins Non-virus antigenic peptides or proteins
  • 2A peptides may also be beneficial when cleavage of the protein of interest from another encoded polypeptide element is desired.
  • 2A peptides may be derived from foot-and-mouth diseases virus, from equine rhinitis A virus, Thosea asigna virus, Porcine teschovirus-l.
  • the artificial nucleic acid sequence particularly the NA sequence according to the inventionD may additionally encode at least one helper peptide.
  • helper peptides binds to class II MHC molecules as a nonspecific vaccine helper epitope (adjuvant) and induces an increased (and long term) immune response by increasing the helper T-cell response.
  • a helper peptide may be N-terminally and/or C-terminally fused to the antigenic peptide or protein derived from Norovirus.
  • any of the above modifications may be applied to the artificial nucleic acid of the present invention, and further to any nucleic acid as used in the context of the present invention and may be, if suitable or necessary, be combined with each other in any combination, provided, these combinations of modifications do not interfere with each other in the artificial nucleic acid.
  • a personD skilled in the art will be able to take his choice accordingly.
  • the artificial nucleic acid as defined herein may preferably comprise a 5'-UTR, a coding region encoding the at least one polypeptide comprising at least one Norovirus protein as described herein, or a fragment, variant or derivative thereof; and/or a 3'-UTR preferably containing at least one histone stem-loop.
  • the 3'-UTR of the artificial nucleic acid preferably comprises also5 a poly(A) and/or a poly(C) sequence as defined herewithin.
  • the single elements of the 3'-UTR may occur therein in any order from 5' to 3' along the sequence of the artificial nucleic acid.
  • further elements as described herein may also be contained, such as a stabilizing sequence as defined herewitbin (e.g.
  • each of the elements may also be repeated in the artificial nucleic acid according to the invention at least once (particularly in di- or multicistronic constructs), preferably twice or more.
  • the single elements may be present in the artificial nucleic acid in the following order:
  • the encoded peptide Dr protein is preferably no histone protein, no reporter protein (e.g. Luciferase, GFP, EGFP, ⁇ -Galactosidase, particularly EGFP) and/or no marker or selection protein (e.g. alpha-Globin, Galactokinase and Xanthine:Guanine phosphoribosyl transferase (GPT)).
  • the artificial nucleic acid according to the invention does not comprise a reporter gene or a marker gene.
  • the artificial nucleic acid according to the invention does not encode, for instance, luciferase; green fluorescent protein (GFP) and its variants (such as eGFP, RFP or FJFP); ot-globin; hypoxanthine-guanine phosphoribosyltransferase (HGPRT); ⁇ -galactosidase; galactokinase; alkaline phosphatase; secreted embryonic alkaline phosphatase (SEAP)) or a resistance gene (such as a resistance gene against neomycin, puromycin, hygramycin and zeocin).
  • the artificial nucleic acid according to the invention does not encode luciferase.
  • the artificial nucleic acid according to the invention does not encode GFP or a variant thereof.
  • the inventive artificial nucleic acid comprises or consists of, preferably in 5' to 3' direction, the following elements:
  • a poly(A) tail preferably consisting of 10 to 20D, ID to IOD, 4D to 8D or 50 to 70 adenosine nucleotides
  • a poly(C) tail preferably consisting of 10 to 200, 10 to IOD, 20 to 70, 20 to B0 or 10 to 40 cytosine nucleotides
  • the artificial nucleic acid according to the invention comprises or consists of, preferably in 5' to 3' direction, the following elements:
  • a 3'-UTR element comprising a nucleic acid sequence, which is derived from an a-globin gene, preferably comprising the corresponding RNA sequence of the nucleic acid sequence according to SEQ ID NO: 39701, or SEQ ID NO: 39702, or a homolog, a fragment or a variant thereof,
  • a poly(A) tail preferably consisting of 10 to 200, 10 to 100, 0 to 80 or 50 to 70 adenosine nucleotides
  • a poly(C) tail preferably consisting of 10 to 200, 10 to 100, 20 to 70, 20 to B0 or 10 to 40 cytosine nucleotides
  • RNA sequence according to the invention comprises, preferably in 5'- to 3'-direction:
  • a) a 5'-cap structure (capO, capl. cap2), preferably m7GpppN;
  • a 5'-0TR element which comprises or consists of a nucleic acid sequence corresponding to a nucleic acid sequence according to SEQ ID NO: 39691, or SEQ ID NO: 39G92, or SEQ ID NO: 39G93, or SEQ ID NO: 39694. a homolog, a fragment or a variant thereof:
  • a 3'-UTR element comprising or consisting of a nucleic acid sequence which is derived from a gene providing a stable RNA, preferably comprising or consisting of the corresponding to a nucleic acid sequence according to SEQ ID NO: 39707, or SEQ ID NO: 39708, or SEQ ID NO: 39703, or SEQ ID NO: 39704, a homolog, a fragment or a variant thereof: e) optionally, a poly(A) sequence preferably comprising 64 adenosines; and
  • f) optionally, a poly(C) sequence, preferably comprising 30 cytosines.
  • the artificial nucleic acid according to the invention comprises or consists of, preferably in 5' to 3' direction, the following elements:
  • a 5'-cap structure (capO, capl, cap2), preferably m7GpppN
  • a 5'-QTR element which comprises or consists of a nucleic acid sequence, which is derived from the 5'-UTR of a TOP gene, preferably comprising a nucleic acid sequence according to SEQ ID NO: 39691 to SED ID NO: 39694, or a homolog, a fragment Dr a variant thereof,
  • a 3'-DTR element comprising a nucleic acid sequencE, which is derived from an albumin gene, preferably comprising the corresponding RNA sequence of the nucleic acid SEquence according to SEO ID NO: 33705, or SEQ ID NO: 3B7DB, or a homolog, a fragment or a variant thereof,
  • the at least one coding region of the artificial nucleic acid according to the present invention comprises a nucleic acid sequence encoding a molecular tag. More preferably, the molecular tag is selected from the group consisting of a FLAG tag, a glutathionE-S-transferase (GST) tag, a His tag, a Myc tag, an E tag, a Strep tag, a green fluorescent protein (GFP) tag and an HA tag.
  • GST glutathionE-S-transferase
  • the mRNA sequBnce according to the invention comprises the following mRNA sequences (or RNA sequences being identical or at least 50%, 60%, 7096. 80%.85%, 86%. 87%, 88%, 8B%, 90%, 91%, 92%, 93%, 94%, 95%, 36%. 97%, 98%, or 99% identical to the following RNA sequences):
  • the artificial nucleic acid according to the invention may be prepared by using any suitable method known in the art, including synthetic methods such as e.g. solid phase synthesis, as well as recombinant and in vitro methods, such as in vitro transcription5 reactions.
  • a linear DNA template is transcribed in vitro using DNA dependent T7 RNA polymerase in the presence of a nucleotide mixture and cap analog (m7GpppG) under suitable buffer conditions.
  • RNA production is performed under current good manufacturing practice, implementing various quality control steps, e.g. accordingD to W0 2DI6/I80430.
  • the obtained RNAs are HPLC purified using PureMessenger 0 (CureVac, Tubingen, Germany; W0 2D08/D77592).
  • purified RNA product is lyophilized according to WD 2DI6/I6583I to yield a temperature stable Norovirus artificial nucleic acid.
  • methods as disclosed in the PCT application PCT/EP20I6/Q82487 are preferably used and adapted accordingly.
  • the present invention provides a composition comprising at least one artificial nucleic acid as described herein and a suitable carrier, preferably a pharmaceutically acceptable carrier.
  • a suitable carrier preferably a pharmaceutically acceptable carrier.
  • the inventive composition comprising the artificial nucleic acid as described herein is preferably a (pharmaceutical) composition or a vaccine as described herein.
  • the inventive composition may comprise either only one type of artificial nucleic acid or at least two different artificial nucleic acids.
  • inventive composition may comprise at least two artificial nucleic acids as described herein, wherein each of the at least two artificial nucleic acids comprises at least one coding region encoding at least one polypeptide comprising a different one of the Norovirus proteins as described herein, or a fragment or a variant of any one of these proteins.
  • the composition may comprise at least two artificial nucleic acids as described herein, wherein each of the at least two artificial nucleic acids comprises at least one coding region encoding at least one polypeptide comprising at least two different Norovirus proteins as described herein, or a fragment or a variant Df any one of these proteins.
  • the composition may also comprise at least two different artificial nucleic acids, which are bi- or multicistronic nucleic acids as described herein and wherein each of the artificial nucleic acids encodes at least two polypeptides, each comprising at least one
  • Norovirus protein or a fragment or variant thereof.
  • the inventive composition comprises or consists of at least one artificial nucleic acid as described herein and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier preferably includes the liquid or non-liquid basis of the inventive composition, which is preferably a pharmaceutical composition or a vaccine. If theD inventive composition is provided in liquid form, the carrier will preferably be water, typically pyrogeo-free water; isotonic saline or buffered (aqueous) solutioos, e.g phosphate, citrate etc. buffered solutions.
  • a buffer more preferably an aqueous buffer, may be used, containing a sodium salt, preferably at least 5D mM of a sodium salt, a calcium salt, preferably at least 0,01 mM of a calcium salt, and optionally a potassium salt, preferably at least 3 mM of a potassium salt.
  • the sodium, calcium and, optionally, potassium salts may occur in the form of their halogenides, e.g.5 chlorides, iodides, or bromides, in the form of their hydroxides, carbonates, hydrogen carbonates, or sulfates, etc.
  • examples of sodium salts include e.g.
  • NaCI, Nal, NaBr, NazC03, NaHC03, NazSOs examples of the optional potassium salts include e.g. KCI, Kl, KBr, K2CO3, KHCO3, 2SO4, and examples of calcium salts include e.g. CaClz. Cab, CaBr2, CaCOs, CaSOi, Ca(DH)z.
  • organic anions of the aforementioned cations may be contained in the boffer. 0
  • one or more compatible solid or liquid fillers or diluents or encapsulating compounds may be used as well, which are suitable for administration to a person.
  • compatible means that the constituents of the inventive composition are capable of being mixed with the at least one artificial nucleic acid of the composition, in such a manner that no interaction occors, which would substantially reduce the biological activity or the pharmaceutical effectiveness of the inventive composition under typical use conditions.
  • Pharmaceutically acceptable carriers, fillers and diluents must, of course, have5 sufficiently high purity and sufficiently low toxicity to make them suitable for administration to a person to be treated.
  • Some examples of compounds which can be used as pharmaceutically acceptable carriers, fillers or constituents thereof are sugars, such as, for example, lactose, glucose, trehalose and sucrose; starches, such as, for example, corn starch or potato starch; dextrose; cellulose and its derivatives, such as, for example, sodium carboxymethylcellulose, ethylcellulose, cellulose acetate; powdered tragacanth; malt; gelatin; tallow; solid gliddants, such as, for example, stearic acid, magnesium stearate; calcium sulfate; vegetable oils, such as, for example, groundnut oil, cottonseed oil, sesame oil, olive oil, corn oil and oil from theobroma; 5 polyols, such as, for example, polypropylene glycol, glycerol, sorbitol, mannitol and polyethylene glycol; alginic acid.
  • sugars such as, for example, lactose, glucose, tre
  • emulsifiers such as, for example, Tween
  • wetting agents such as, far example, sodium lauryl sulfate
  • colouring agents such as, far example, sodium lauryl sulfate
  • taste-imparting agents pharmaceutical carriers
  • tablet-forming agents such as, butyl sulfate
  • antioxidants such as, sodium lauryl sulfate
  • the composition of the invention comprises at least 2, 3, 4, 5, 6, 7, 8, 3, ID, II, 12, 13, 14. 15, IB, 17, 18, 13, 20, 1, 22.23, 24, 25, 2B, 27, 28, 23, 30, 31. 32, 33, 34, 35, 3B, 37, 38, 33, 40, 41, 42, 3, 44, 45, B, 47, 48, 43, 50, 51, 52, 53, 54, 55, 5B, 57, 58, 53, GO, Gl, G2, 63, G4, 65, 66, 67.
  • each coding region encoding at least one polypeptide comprising a Norovirus protein, and/ or a fragment or a variant of any one of these proteins, wherein each coding region preferably encodes a different Norovirus protein, more0 preferably each coding region encodes a capsid protein, preferably VPI of a different Norovirus.
  • the composition of the invention comprises at least 2, 3, 4, 5, B, 7, 8, 9, 10, II, 12, 13, 14, 15, IB, 17, 18, 19, 20, 21, 22, 23, 24, 25,26, 7, 28, 29.30, 31.32. 33, 34, 35, 36, 37, 38, 39, 40, 41, 42.43.44, 45, 46, 47, 48, 49, 5051, 52, 53.54, 55, 56, 57, 58, 59, B0, 61, 62.63, 64, 65, B6, B7, 68, B9, 70.71, 72, 73, 74, 75, 76, 77, 78.79, 8D.
  • artificial nucleic acids of the invention comprises at least one coding region encoding at least one polypeptide comprising at least two different Norovirus proteins.0 preferably VPI and VP2, and/ or a fragment or a variant of any one of these proteins.
  • the inventive composition which is preferably a pharmaceutical composition or a vaccine, comprises at least one artificial nucleic acid as described herein, wherein the at least one artificial nucleic acid is complexed at least partially with a cationic Dr polycationic compound and/or a polymeric carrier, preferably a cationic protein or peptide.5
  • the at least one artificial nucleic acid as defined herein or any other nucleic acid comprised in the inventive (pharmaceutical) composition or vaccine is associated with or complexed with a cationic or polycationic compound or a polymeric carrier, optionally in a weight ratio selected from a range of about 6:1 (w/w) to about 0.25:1 (w/w), more preferably from about 5:1 (w/ w) to about 0.5:1 (w/ w), even more preferably of about 4:1 (w/w) to about 1:1 (w/w) or of about 3:1 (w/w) to about 1:1 (
  • the N/P ratio of the at least one artificial nucleic acid to the one or mure polycations is in the range of about 0.1 to 10, including a range of about 0.3 to 4, of about 0.5 to 2, of about 0.7 to 2 aod of about 0.7 to 1.5.
  • the composition comprising at least one artificial nucleic acid of the invention is defined as follows: (i) the ratio of complexed nucleic acid to free nucleic acid is selected from a range of about 5:1 (w/w) to about 1:10 (w/w), more preferably from a range of about 4:1 (w/w) to about 1:8 (w/w), even more preferably from a range of about 3:1 (w/w) to about 1:5 (w/w) or 1:3 (w/w), wherein the ratio is most preferably about 1:1 (w/w); or
  • the mRNA is complexed with one or more cationic or polycationic compounds in a weight ratio selected from a range of about B:l (w/w) to about 0.25:1 (w/w), more preferably from about 5:1 (w/w) to about 0.5:1 (w/w), even more preferably of about 4:1 (w/w) to about 1:1 (w:w) or of about 3:1 (w/w) to about 1:1 (w/w), and most preferably a ratio of about 3:1 (w/w) to about 2:1 (w/w) of mRNA to cationic or polycationic compound and/or with a polymeric carrier; or optionally in a nitrogen/phosphate ratio of mRNA to cationic or polycationic compound and/or polymeric carrier in the range of about 0.1-10. preferably in a range of about 0.3-4 or 0.3-1, and most preferably in a range of about 0.5-1 or 0.7-1, and even most preferably in a range of about 0.3-O.B or 0.5
  • the at least one artificial nucleic acid or mRNA is complexed with one or more cationic or polycationic compounds, preferably with cationic or polycationic polymers, cationic or polycationic peptides or proteins, e.g. protamine, cationic or polycationic polysaccharides and/or cationic or polycationic lipids and/or wherein the at least one artificial nucleic acid or mRNA is complexed with one or more lipids and thereby forming liposomes, lipid nanoparticles and/or lipoplexes.
  • cationic or polycationic compounds preferably with cationic or polycationic polymers, cationic or polycationic peptides or proteins, e.g. protamine, cationic or polycationic polysaccharides and/or cationic or polycationic lipids and/or wherein the at least one artificial nucleic acid or mRNA is complexed with one or more lipids and thereby forming liposomes
  • the inventive composition comprises at least one artificial nucleic acid as described herein, which is complexed with one or more polycations and/or a polymeric carrier, and at least one free nucleic acid, wherein the at least one complexed nucleic acid is preferably identical to the at least one artificial nucleic acid according to the present invention.
  • the at least one artificial nucleic acid of the inventive composition is complexed at least partially with a cationic or polycationic compound and/or a polymeric carrier, preferably cationic proteins or peptides.
  • W02010/037539 and W02012/113513 is incorporated herewith by reference.
  • composition of the invention comprises
  • compositions and/or vaccines of the invention comprise artificial nucleic acids encoding one or more capsid proteins VPI derived from one or more Noroviruses. In another embodiment, the compositions and/or vaccines of the invention comprise artificial nucleic acids encoding one or more capsid proteins VP2 derived from one or more Noroviruses.
  • composition of the invention is further defined as composition, wherein
  • the artificial nucleic acids are derived from a single Gl Norovirus or from 2, 3, , 5, G, 7, 8, 9, 10, II, 12.13, 14, 15, IB, 17, 18, 19, 20, 21, 22, 23, 24, 25, 2G, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, G, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, G4, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80.
  • the artificial nucleic acids are derived from a single Gil Norovirus or from 2, 3.4, 5, 6, 7. 8, 9, 10, II, 12, 13, 14, 15, IB, 17, 18, 19, 20, 21, 22, 3, 4, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 6, 47, 48, 49, 50, 51, 52, 53, 54, 55.
  • the artificial nucleic acids are derived from a single Gill Norovirus or from 2, 3, , 5, 6.7, 8, 9, 10, II, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23.24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 4D, 41, 2, 43, 44, 45, 4B, 47, 8, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, G6, B7, 68, 69, 70.71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, SB. 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 10D or more different Gill Noroviruses; or
  • the artificial nucleic acids are derived from a single GIV Norovirus or from 2, 3, 4, 5, 6, 7, 8, 9, 10, II, 12, 13, 14. 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 3D. 31, 32, 33, 34, 35, 3B, 37, 38, 39, 40, 41, 42, 43, 44. 45, 6, 7, 48, 9, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, Gl, 62.
  • the artificial nucleic acids are derived from a single GV Norovirus or from 2, 3, 4, 5, G, 7, 8, 9, 10, II, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 4, 25, 26, 27, 28, 29, 3D, 31, 32, 33, 34, 35, 36, 37, 38, 39, 4D, 41, 42. 43. 44, 45, 6, 47, 8, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85. 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 10D or more different GV Noroviruses; or
  • the artificial nucleic acids are derived from a single Gl Norovirus and additionally from a single Gil Norovirus, Gill Norovirus, GIV Norovirus and/or GV Norovirus; or
  • the artificial nucleic acids are derived from a single Gl Norovirus or from 2, 3, 4, 5, 6, 7, 8, 9, 10, II, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 4, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, G, 47.
  • composition of the invention further is defined as composition, wherein
  • the artificial nucleic acids are derived from a single Gl.l Norovirus or from 2, 3, , 5, 6, 7, 8, 9, ID, II, 12, 13, 14, 15, 16, 17, 18, 19, 2D, 21, 22, 23, 24, 25, 26. 27, 28, 29, 3D, 31. 32, 33, 34, 35, 36, 37, 38, 39, 4D, 41, 2, 43, 44, 45, 6, 47, 48, 49, 50. 51, 52, 53, 54, 55, 56, 57. 58, 59, 60, 61, 62, 63, 64, 65, 66. 67, 68, 69, 7D, 71, 72, 73, 74, 75, 76, 77, 78, 79, 8D, 81,
  • the artificial nucleic acids are derived from a single GII.4 Norovirus or from 2, 3, , 5, 6, 7, 8, 8, ID, II, 12, 13, 14, 15, 16, 17, 18, 19, 2D, 21, 22, 23, 24, 25, 26, 27, 28.20, 30.31.32, 33, 34, 35, 36, 37, 38, 30, 40, 41, 42, 43.44, 45, 46.47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 50, 60, 61, 62, 63, 64, 65, 66, 67, 68, 60, 70, 71, 72. 73.74, 75, 76, 77, 78, 70.
  • the artificial nucleic acids are derived from a single Gl.l Norovirus and additionally from a single GII.4 Norovirus; or
  • the artificial nucleic acids are derived from a single Gl.l Norovirus or from 2, 3, 4, 5, 6, 7, 8, 0, 10, II, 12, 13, 14, 15, 16, 17, 18, 10, 20, 21, 22, 23, 24, 25, 26. 27, 28, 20, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 6, 47, 48, 49. 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84. 85.
  • Gl.l Noroviruses and additionally from a single GII.4 Norovirus or from 2, 3, 4, 5, 6, 7, 8, 9, 10. II, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 5D, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61. 62. 63, 64.
  • composition of the invention further is defined as composition, wherein
  • the artificial nucleic acids are derived from a single Gl.l Norovirus or from 2, 3, 4, 5, 6, 7, 8, 0, ID, II, 12, 13, 14, 15, 16, 17, 18, 19, 2D, 21, 22, 23, 24, 25, 26, 27, 28, 20, 30, 31, 32. 33, 34, 35, 36, 37, 38, 30, 0, 41, 42, 43, 44, 45, 46, 47, 8, 49, 5D. 51, 52, 53, 54, 55, 56. 57.
  • the artificial nucleic acids are derived from a single GII.4 Norovirus or from 2, 3, 4, 5, 6, 7, 8, 9, ID, II, 12, 13, 14, 15. 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 3, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 or more different GII.4 Noroviruses; or (iii) the artificial nucleic acids are derived from 2, 3,
  • Gl.l Noroviruses and additionally from a single GII.4 Norovirus or from 2. 3. , 5, 6.7, 8, 9, 10, II, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27. 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50.
  • nucleic acid sequences having, in increasing order of preference, at least 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 7096. 71%. 72%.73%. 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%. 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to the nucleic acid sequence represented by any one of SEO ID NOs: 4411-39690, 39713-39746; and/or
  • nucleic acid sequences having, in increasing order of preference, at least 50%, 51%, 52%, 53%, 54%, 55%, 56%. 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%. 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%.
  • the composition of the invention further is defined as a composition comprising two different nucleic acid sequences (bivalent, divalent composition), wherein one of the two different nucleic acid sequences is derived from Norovirus GII.4 and one of the two different nucleic acid sequence is derived from Norovirus Gl.l, wherein the one nucleic acid sequence of Norovirus GII.4 may be any one of the nucleic acid sequences as defined herein and in Table I, or fragment or variants of these sequences, and wherein the one nucleic acid sequence of Noravirus Gl.l may be any one of the nucleic acid sequences as defined herein and in Table I, or fragment or variants of these sequences.
  • the composition comprising two different nucleic acid sequences comprises or consists of one nucleic acid sequence derived from Nurovirus GII.4 selected from SED ID NDs: 39713-39721, 39726-39742, and one nucleic acid sequence derived from Noravirus Gl.l 5 selected from SED ID NDs: 39722-39725.
  • the composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid sequences SED ID ND: 397IG (Norovirus GII.4) and SEQ ID NO: 39725 (Norovirus Gl.l).
  • the composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid sequences SED ID ND: 397IG (Norovirus GII.4) and SEQ ID NO: 39725 (Norovirus Gl.l).
  • the composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid sequences SED ID ND: 397IG (Norovirus GII.4) and SEQ ID NO: 39725 (Norovirus Gl.l).
  • composition ID comprises or consists of the nucleic acid sequences SED ID ND: 33721 (Norovirus GII.4) and SED ID ND: 39725 (Norovirus Gl.l).
  • composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid sequences SED ID ND: 39729 (Norovirus GII.4) and SED ID ND: 39725 (Norovirus Gl.l).
  • composition comprising two different nucleic acid sequences (bivalent, divalent composition) comprises or consists of the nucleic acid sequences SEQ ID ND: 39734
  • composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid sequences SED ID ND: 39738 (Norovirus GII.4) and SED ID ND: 39725 (Norovirus Gl.l).
  • the composition of the invention further is defined as a composition comprising two different nocleicD acid sequences (bivalent, divalent composition) derived from Norovirus GII.4, wherein the two different nucleic acid sequences of Norovirus GII.4 may be any one of the nucleic acid sequences as defined herein and in Table I, or fragment or variants of these sequences.
  • the composition comprising two different nucleic acid sequences derived from Norovirus GII.4 comprises or consists of one nucleic acid sequence selected from SED ID NDs: 39713-39721, 39726-39742.
  • the composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid sequences SED ID ND: 39716 (Norovirus GII.4) and SEQ ID ND: 39721 (Norovirus GII.4).
  • the composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid sequences SEQ ID ND: 39729 (Norovirus GII.4) and SEQ ID ND: 39721 (Norovirus GII.4).
  • the composition comprising two different nucleic acid sequencesD comprises or consists of the nucleic acid sequences SED ID NO: 39734 (Norovirus GII.4) and SED ID ND: 39721 (Norovirus GII.4).
  • the composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid sequences SED ID ND: 39738 (Norovirus GII.4) and SED ID ND: 39721 (Norovirus GII.4).
  • the composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid sequences SEQ ID ND: 397165 (Norovirus GII.4) and SED ID NO: 39729 (Norovirus GII.4).
  • the composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid seqoences SEQ ID ND: 39734 (Norovirus GII.4) and SEQ ID NO: 39729 (Norovirus GII.4).
  • the composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid sequences SEfl ID NO: 38738 (Norovirus GII.4) and SEO ID NO: 39729 (Norovirus GII.4).
  • the composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid sequences SEO 10 NO: 397IG (Norovirus GII.4) and SEQ ID NO: 39734 (Norovirus Gll.4).ln a specific preferred embodiment, the composition comprising two different nucleic acid sequences (bivalent, divalent composition) comprises or consists of the nucleic acid sequences SED ID NO: 39738 (Norovirus GII.4) and SEfl ID NO: 39734 (Norovirus GII.4).
  • the composition comprising two different nucleic acid sequences comprises or consists of the nucleic acid sequences SEfl ID NO: 397IG (Norovirus GII.4) and SEfl ID NO: 39738 (Norovirus GII.4).
  • the composition of the invention further is defined as a composition comprising four different nucleic acid sequences (tetravalent composition), wherein each of the four different nucleic acid sequences is derived from Norovirus GII.4, wherein each of the four different nucleic of Norovirus GII.4 may be any one of the nucleic acid sequences as defined herein and in Table I, or fragment or variants of these sequences.
  • the composition comprising four different nucleic acid sequences (tetravalent composition) comprises or consists of four nucleic acid sequence derived from Norovirus GII.4 selected from SEfl ID NOs: 39713-39721, 3972G-39742.
  • the composition comprising four different nucleic acid sequences comprises four of the nucleic acid sequences selected from SEQ ID NOs: 39716, 39721, 39729, 39734 or 39738.
  • the composition of the invention is defined as a composition comprising four different nucleic acid sequences (tetravalent composition), wherein at least one of the four different nucleic acid sequences is derived from Norovirus GII.4 and at least one of the four different nucleic acid sequence is derived from Norovirus Gl.l, wherein the at least one of the four different nucleic of Norovirus GII.4 may be any one of the nucleic acid sequences as defined herein and in Table I, or fragment or variants of these sequences, and wherein the at least one of the four of nucleic acid sequence of Norovirus Gl.l may be any one of the nucleic acid sequences as defined herein and in Table I, or fragment or variants of these sequences.
  • the composition of the invention is defined as a composition comprising four different nucleic acid sequences (tetravalent composition), wherein three of the four different nucleic acid sequences are derived from Norovirus GII.4 and one of the four different nucleic acid sequence is derived from Norovirus Gl.l, wherein the three of the four different nucleic of Norovirus GII.4 may be any one of the nucleic acid sequences as defined in Table I, or fragment or variants of these sequences, and wherein the one of the four of nucleic acid sequence of Norovirus Gl.l may be any one of the nucleic acid sequences as defined in Table I, or fragment or variants of these sequences.
  • the composition comprising four different nucleic acid sequences comprises or consists of three nucleic acid sequence derived from Norovirus GII.4 selected from SEfl ID NOs: 39713-39721, 3972B-39742 and one nucleic acid sequence derived from Norovirus Gl.l SEQ ID NOs: 39722-39725.
  • the composition comprising four different nucleic acid sequences comprises three of the nucleic acid sequences derived from Norovirus GII.4 selected from SEQ ID NOs: 397I6. 3972I, 3372B, 39734 or 39738 and one nucleic acid sequence derived from Norovirus Gl.l SEQ ID NO: 39725.
  • the composition of the invention is defined as a composition comprising multiple different nucleic acid sequences (multivalent composition) defined as a composition comprising 5, G, 7, 8, 9, ID, II, I2, 13, 14, 15, IB, I7, 18, I9, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31. 32, 33, 34, 35, 36. 37, 38, 39, 4Q, 41, 42, 43. 44, 45, 4G, 47, 48, 49, 50, 51, 52.
  • the composition of the invention is defined as a composition comprising multiple different nucleic acid sequences (multivalent composition) defined as a composition comprising 5, 6, 7, 8, 9,I0, II, I2, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 7, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 0, 1, 2, 43, 4, 45, 46.47, 48, 49, 5D, 51, 52, 53, 54, 55, 56.
  • multivalent composition defined as a composition comprising 5, 6, 7, 8, 9,I0, II, I2, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 7, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 0, 1, 2, 43, 4, 45, 46.47, 48, 49, 5D, 51, 52, 53, 54, 55, 56.
  • each row corresponds to selected Norovirus protein or antigen as identified by the respective name (first column, column I "Strain/Isolate”) and the database accession number of the corresponding protein (second column, column 2 "NC6I or Genbank Accession No.”).
  • the third column, column 3 ("A") in Table 3 indicates the SEQ ID NOs corresponding to the respective amino acid sequence as provided herein.
  • the SEQ ID NOs corresponding to the nucleic acid sequence of the wild5 type nucleic acid sequence encoding the Norovirus protein or peptide is indicated in the fourth column, column 4 ("B").
  • the fifth column, column 5 provides the SEQ ID NOs corresponding to modified nucleic acid sequences of the nucleic acid sequences as described herein that encode the Norovirus protein or peptide preferably having the amino acid sequence as defined by the SEQ ID NOs indicated in the third column (“A”) or by the database entry indicated in the second column ("NCBI or Genbank Accession No.”).
  • Narovirus AII737I7 877 5287 9697,14107,18517, 22927,

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Dermatology (AREA)
  • Immunology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention concerne un acide nucléique artificiel et des polypeptides appropriés pour une utilisation dans le traitement ou la prophylaxie d'une infection par un novovirus ou d'un trouble lié à une telle infection. En particulier, la présente invention concerne un vaccin contre un novovirus. La présente invention concerne un acide nucléique artificiel, des polypeptides, des compositions et des vaccins comprenant l'acide nucléique artificiel ou les polypeptides. La présente invention concerne en outre une méthode de traitement ou de prévention d'un trouble ou d'une maladie, des première et seconde utilisations médicales de l'acide nucléique artificiel, des polypeptides, des compositions et des vaccins. Enfin, la présente invention concerne un kit, en particulier un kit d'éléments, comprenant l'acide nucléique artificiel, les polypeptides, les compositions et les vaccins.
EP17725524.7A 2016-05-04 2017-05-04 Molécules d'acide nucléique et leurs utilisations Pending EP3452493A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP2016060115 2016-05-04
PCT/EP2017/060673 WO2017191264A1 (fr) 2016-05-04 2017-05-04 Molécules d'acide nucléique et leurs utilisations

Publications (1)

Publication Number Publication Date
EP3452493A1 true EP3452493A1 (fr) 2019-03-13

Family

ID=58772538

Family Applications (1)

Application Number Title Priority Date Filing Date
EP17725524.7A Pending EP3452493A1 (fr) 2016-05-04 2017-05-04 Molécules d'acide nucléique et leurs utilisations

Country Status (3)

Country Link
US (2) US11141474B2 (fr)
EP (1) EP3452493A1 (fr)
WO (1) WO2017191264A1 (fr)

Families Citing this family (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012116714A1 (fr) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination chez des patients âgés
MX2015010880A (es) 2013-02-22 2015-12-03 Curevac Gmbh Combinacion de vacunacion e inhibicion de la trayectoria pd-1.
AU2014310931B2 (en) 2013-08-21 2019-12-19 CureVac SE Rabies vaccine
MX369469B (es) 2013-08-21 2019-11-08 Curevac Ag Vacuna contra el virus respiratorio sincitial.
ES2946969T3 (es) 2014-12-12 2023-07-28 CureVac SE Moléculas de ácido nucleico artificiales para una expresión proteica mejorada
DK3294885T3 (da) 2015-05-08 2020-08-10 Curevac Real Estate Gmbh Fremgangsmåde til at fremstille rna
EP4108769B1 (fr) 2015-05-29 2023-08-30 CureVac Manufacturing GmbH Procédé de production et de purification de l'arn, comprenant au moins une étape de filtration à flux tangentiel
US9765117B2 (en) * 2015-08-24 2017-09-19 Romek Figa Peptides for treating cancer
US11225682B2 (en) 2015-10-12 2022-01-18 Curevac Ag Automated method for isolation, selection and/or detection of microorganisms or cells comprised in a solution
US11413346B2 (en) 2015-11-09 2022-08-16 Curevac Ag Rotavirus vaccines
DK3319622T3 (da) 2015-12-22 2020-05-04 Curevac Ag Fremgangsmåde til fremstilling af rna-molekylesammensætninger
WO2017109161A1 (fr) 2015-12-23 2017-06-29 Curevac Ag Procédé de transcription in vitro d'arn utilisant un tampon contenant un acide dicarboxyliqlue ou un acide tricarboxylique ou un sel de celui-ci
BR112018016755A2 (pt) 2016-02-17 2018-12-26 Curevac Ag vacina contra o vírus da zica
US11920174B2 (en) 2016-03-03 2024-03-05 CureVac SE RNA analysis by total hydrolysis and quantification of released nucleosides
WO2017186928A1 (fr) 2016-04-29 2017-11-02 Curevac Ag Arn codant pour un anticorps
WO2017191264A1 (fr) * 2016-05-04 2017-11-09 Curevac Ag Molécules d'acide nucléique et leurs utilisations
US11078247B2 (en) 2016-05-04 2021-08-03 Curevac Ag RNA encoding a therapeutic protein
US11478552B2 (en) 2016-06-09 2022-10-25 Curevac Ag Hybrid carriers for nucleic acid cargo
US11279923B2 (en) 2016-11-28 2022-03-22 Curevac Ag Method for purifying RNA
US11464836B2 (en) 2016-12-08 2022-10-11 Curevac Ag RNA for treatment or prophylaxis of a liver disease
WO2018104540A1 (fr) 2016-12-08 2018-06-14 Curevac Ag Arn pour la cicatrisation des plaies
US11141476B2 (en) 2016-12-23 2021-10-12 Curevac Ag MERS coronavirus vaccine
WO2018115507A2 (fr) 2016-12-23 2018-06-28 Curevac Ag Vaccin contre l'hénipavirus
WO2018115525A1 (fr) 2016-12-23 2018-06-28 Curevac Ag Vaccin contre le virus de lassa
SG10202110491PA (en) 2017-03-24 2021-11-29 Curevac Ag Nucleic acids encoding crispr-associated proteins and uses thereof
WO2018191657A1 (fr) 2017-04-13 2018-10-18 Acuitas Therapeutics, Inc. Lipides pour administration d'agents actifs
EP3424524A3 (fr) 2017-07-04 2019-02-27 CureVac AG Nouvelles molécules d'acide nucléique
US11602557B2 (en) 2017-08-22 2023-03-14 Cure Vac SE Bunyavirales vaccine
JP2021502079A (ja) 2017-11-08 2021-01-28 キュアバック アーゲー RNA配列の適合(Adaptation)
KR20200092363A (ko) * 2017-11-30 2020-08-03 메디카고 인코포레이티드 변형된 노로바이러스 vp1 단백질 및 변형된 노로바이러스 vp1 단백질을 포함하는 vlp
WO2019115635A1 (fr) 2017-12-13 2019-06-20 Curevac Ag Vaccin contre les flavivirus
WO2019122371A1 (fr) 2017-12-21 2019-06-27 Curevac Ag Adn linéaire double brin couplé à un support ou une étiquette unique et procédés de production dudit adn linéaire double brin
JP7429684B6 (ja) 2018-07-13 2024-03-15 メディカゴ インコーポレイテッド 改変ノロウイルスvp1タンパク質および改変ノロウイルスvp1タンパク質を含むvlp
CN109180810B (zh) * 2018-09-27 2021-05-07 国药中生生物技术研究院有限公司 特异性结合诺如病毒gi.1基因型vp1蛋白或vlp的抗体及其制备方法和应用
EP3673917A1 (fr) * 2018-12-28 2020-07-01 Themis Bioscience GmbH Vaccins de norovirus
CN110176976B (zh) * 2019-06-28 2021-02-09 中车青岛四方车辆研究所有限公司 列车时间校准系统、方法及时间校准设备
US20240277830A1 (en) 2020-02-04 2024-08-22 CureVac SE Coronavirus vaccine
US11241493B2 (en) 2020-02-04 2022-02-08 Curevac Ag Coronavirus vaccine
JP2024502210A (ja) 2020-12-22 2024-01-17 キュアバック エスイー SARS-CoV-2バリアントに対するRNAワクチン
CA3235832A1 (fr) * 2021-10-27 2023-05-04 Elena ATOCHINA-VASSERMAN Vaccin contre un norovirus et methodes d'utilisation
WO2024015890A1 (fr) * 2022-07-13 2024-01-18 Modernatx, Inc. Vaccins à arnm de norovirus
WO2024144272A1 (fr) * 2022-12-27 2024-07-04 (주)한국바이오셀프 Composition vaccinale comprenant de l'arnm de norovirus gii
WO2024144262A1 (fr) * 2022-12-27 2024-07-04 (주)한국바이오셀프 Composition de vaccin comprenant de l'arnm de norovirus gi
CN118126202B (zh) * 2024-05-07 2024-07-30 上海瑞宙生物科技有限公司 一种诺如病毒类病毒颗粒及其应用

Family Cites Families (103)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2238799T3 (es) 1999-09-09 2005-09-01 Curevac Gmbh Transferencia de arn-m.
ES2340532T3 (es) 2001-06-05 2010-06-04 Curevac Gmbh Arnm con un contenido g/c aumentado que codifica para un antigeno bacteriano y utilizacion del mismo.
DE10162480A1 (de) 2001-12-19 2003-08-07 Ingmar Hoerr Die Applikation von mRNA für den Einsatz als Therapeutikum gegen Tumorerkrankungen
US7074596B2 (en) 2002-03-25 2006-07-11 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Synthesis and use of anti-reverse mRNA cap analogues
DE10229872A1 (de) 2002-07-03 2004-01-29 Curevac Gmbh Immunstimulation durch chemisch modifizierte RNA
DE10335833A1 (de) 2003-08-05 2005-03-03 Curevac Gmbh Transfektion von Blutzellen mit mRNA zur Immunstimulation und Gentherapie
DE102004042546A1 (de) 2004-09-02 2006-03-09 Curevac Gmbh Kombinationstherapie zur Immunstimulation
DE102005023170A1 (de) 2005-05-19 2006-11-23 Curevac Gmbh Optimierte Formulierung für mRNA
ES2514316T3 (es) * 2005-11-22 2014-10-28 Novartis Vaccines And Diagnostics, Inc. Partículas similares a virus (VLPs) de Norovirus y Sapovirus
CA2659301A1 (fr) 2006-07-28 2008-02-07 Applera Corporation Analogues de coiffes de nucleotide d'arnm
EP2046954A2 (fr) 2006-07-31 2009-04-15 Curevac GmbH Acide nucléique de formule (i): gixmgn, ou(ii): cixmcn, en particulier en tant qu'agent/adjuvant immunostimulant
DE102006035618A1 (de) 2006-07-31 2008-02-07 Curevac Gmbh Nukleinsäure der Formel (I): GlXmGn, insbesondere als immunstimulierendes Adjuvanz
DE102006061015A1 (de) 2006-12-22 2008-06-26 Curevac Gmbh Verfahren zur Reinigung von RNA im präparativen Maßstab mittels HPLC
DE102007001370A1 (de) 2007-01-09 2008-07-10 Curevac Gmbh RNA-kodierte Antikörper
PT2167523E (pt) 2007-06-19 2014-09-22 Univ Louisiana State Síntese e utilização de análogos fosforotiolato antireversos do capuz de arn mensageiro
WO2009030254A1 (fr) 2007-09-04 2009-03-12 Curevac Gmbh Complexes d'arn et de peptides cationiques pour transfection et immunostimulation
WO2009046739A1 (fr) 2007-10-09 2009-04-16 Curevac Gmbh Composition pour traiter le cancer de la prostate (pca)
WO2009046738A1 (fr) 2007-10-09 2009-04-16 Curevac Gmbh Composition pour traiter le cancer du poumon, notamment les cancers du poumon non à petites cellules (nsclc)
MX2010008468A (es) 2008-01-31 2010-08-30 Curevac Gmbh Acidos nucleicos de la formula (i) (nug1xmgnnv)a y derivados de los mismos como un agente/adyuvante inmunoestimulante.
US8023266B2 (en) 2008-05-20 2011-09-20 Greenray Inc. AC photovoltaic module and inverter assembly
WO2010037408A1 (fr) 2008-09-30 2010-04-08 Curevac Gmbh Composition comprenant un arnm complexé et un arnm nu pour déclencher ou augmenter une réponse immunostimulante chez un mammifère et utilisations de ladite composition
CA2766907A1 (fr) * 2009-07-06 2011-01-13 Novartis Ag Molecules d'arn autorepliquantes et leurs utilisations
EP2281579A1 (fr) 2009-08-05 2011-02-09 BioNTech AG Composition de vaccin comportant un ADN modifié 5'-Cap
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
WO2011069529A1 (fr) 2009-12-09 2011-06-16 Curevac Gmbh Solution contenant du mannose pour la lyophilisation, la transfection et/ou l'injection d'acides nucléiques
EP2590670B1 (fr) * 2010-07-06 2017-08-23 GlaxoSmithKline Biologicals SA Procédé pour induire une réponse immunitaire par administration d'arn
CN103025876A (zh) 2010-07-30 2013-04-03 库瑞瓦格有限责任公司 用于转染和免疫刺激的核酸与二硫化物交联的阳离子成分的复合体
WO2012019630A1 (fr) 2010-08-13 2012-02-16 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour augmenter l'expression d'une protéine codée
WO2012089225A1 (fr) 2010-12-29 2012-07-05 Curevac Gmbh Combinaison de vaccination et d'inhibition de la présentation des antigènes restreinte par le cmh de classe i
WO2012116715A1 (fr) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination chez des nouveaux-nés et des enfants en bas âge
WO2012113413A1 (fr) 2011-02-21 2012-08-30 Curevac Gmbh Composition de vaccin comprenant des acides nucléiques immunostimulateurs complexés et des antigènes emballés avec des conjugués de polyéthylèneglycol/peptide à liaison disulfure
WO2012116714A1 (fr) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination chez des patients âgés
KR101275225B1 (ko) 2011-10-17 2013-06-17 엔젯 주식회사 정전기력을 이용한 잉크토출장치
WO2013113326A1 (fr) 2012-01-31 2013-08-08 Curevac Gmbh Composition pharmaceutique comprenant un complexe support polymère - charge et au moins un antigène de protéine ou de peptide
EP2623121A1 (fr) 2012-01-31 2013-08-07 Bayer Innovation GmbH Composition pharmaceutique comportant un complexe de chargement de porteur polymérique et un antigène
WO2013113325A1 (fr) 2012-01-31 2013-08-08 Curevac Gmbh Complexes chargés négativement comprenant des acides nucléiques pour l'immunostimulation
WO2013120499A1 (fr) 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour augmenter l'expression d'un antigène pathogène codé
WO2013120497A1 (fr) 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour l'augmentation de l'expression d'une protéine thérapeutique codée
WO2013120498A1 (fr) 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour augmenter l'expression d'un autoantigène auto-immun ou d'un antigène allergène codé
WO2013120500A1 (fr) 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation en vue d'augmenter l'expression d'un antigène tumoral codé
US9683233B2 (en) 2012-03-27 2017-06-20 Curevac Ag Artificial nucleic acid molecules for improved protein or peptide expression
US10080809B2 (en) 2012-03-27 2018-09-25 Curevac Ag Artificial nucleic acid molecules comprising a 5′TOP UTR
WO2013143698A1 (fr) 2012-03-27 2013-10-03 Curevac Gmbh Molécules d'acide nucléique artificielles
EP3498267A1 (fr) 2012-05-25 2019-06-19 CureVac AG Immobilisation réversible et/ou libération contrôlée d'acide nucléique contenant des nanoparticules par des revêtements polymères (biodégradables)
MX2015010880A (es) 2013-02-22 2015-12-03 Curevac Gmbh Combinacion de vacunacion e inhibicion de la trayectoria pd-1.
MX369469B (es) 2013-08-21 2019-11-08 Curevac Ag Vacuna contra el virus respiratorio sincitial.
WO2015024667A1 (fr) 2013-08-21 2015-02-26 Curevac Gmbh Procédé pour augmenter l'expression de protéines codées par l'arn
KR20160043103A (ko) 2013-08-21 2016-04-20 큐어백 아게 전립선암 치료를 위한 조성물 및 백신
CA2915730A1 (fr) 2013-08-21 2015-02-26 Karl-Josef Kallen Vaccin combine contre le virus respiratoire syncytial (rsv) et la grippea
AU2014310931B2 (en) 2013-08-21 2019-12-19 CureVac SE Rabies vaccine
BR112016003400A2 (pt) 2013-08-21 2017-12-05 Curevac Ag composição e vacina para tratamento de câncer de pulmão
WO2015062738A1 (fr) 2013-11-01 2015-05-07 Curevac Gmbh Arn modifié à propriétés immunostimulantes réduites
SG11201604198YA (en) 2013-12-30 2016-07-28 Curevac Ag Methods for rna analysis
WO2015101415A1 (fr) 2013-12-30 2015-07-09 Curevac Gmbh Molécules d'acides nucléiques artificielles
CN111304231A (zh) 2013-12-30 2020-06-19 库瑞瓦格股份公司 人工核酸分子
WO2015130584A2 (fr) * 2014-02-25 2015-09-03 Merck Sharp & Dohme Corp. Adjuvants de vaccins sous forme de nanoparticules lipidiques et systèmes d'administration d'antigènes
EP3116535B1 (fr) 2014-03-12 2019-08-07 CureVac AG Combinaison de vaccination et d'agonistes de ox40
EP3129050A2 (fr) 2014-04-01 2017-02-15 CureVac AG Complexe cargo de support polymère à utiliser comme agent immunostimulant ou comme adjuvant
WO2015188933A1 (fr) 2014-06-10 2015-12-17 Curevac Ag Procédés et moyen d'amélioration de la production d'arn
ES2946969T3 (es) 2014-12-12 2023-07-28 CureVac SE Moléculas de ácido nucleico artificiales para una expresión proteica mejorada
CA2962849A1 (fr) 2014-12-16 2016-06-23 Curevac Ag Vaccins contre le virus ebola et le virus marburg
JP6907116B2 (ja) 2014-12-30 2021-07-21 キュアバック アーゲー 人工核酸分子
WO2016165825A1 (fr) 2015-04-13 2016-10-20 Curevac Ag Procédé de production de compositions d'arn
EP4026568A1 (fr) 2015-04-17 2022-07-13 CureVac Real Estate GmbH Lyophilisation de l'arn
ES2753259T3 (es) 2015-04-22 2020-04-07 Curevac Ag Composición que contiene ARN para el tratamiento de enfermedades tumorales
CN108064307A (zh) 2015-04-30 2018-05-22 库瑞瓦格股份公司 使用固定化的限制酶进行体外转录的方法
WO2016174271A1 (fr) 2015-04-30 2016-11-03 Curevac Ag Poly(n)polymérase immobilisée
DK3294885T3 (da) 2015-05-08 2020-08-10 Curevac Real Estate Gmbh Fremgangsmåde til at fremstille rna
US11559570B2 (en) 2015-05-15 2023-01-24 CureVac SE Prime-boost regimens involving administration of at least one mRNA construct
CN107530448A (zh) 2015-05-20 2018-01-02 库瑞瓦格股份公司 包含长链rna的干粉组合物
EP3298142B1 (fr) 2015-05-20 2021-07-14 CureVac AG Composition de poudre sèche comprenant de l'arn à chaîne longue
EP3303575B1 (fr) 2015-05-29 2022-03-16 CureVac AG Procédé d'ajout de structures de coiffe à un arn au moyen d'enzymes immobilisées
EP4108769B1 (fr) 2015-05-29 2023-08-30 CureVac Manufacturing GmbH Procédé de production et de purification de l'arn, comprenant au moins une étape de filtration à flux tangentiel
US20180296663A1 (en) 2015-06-17 2018-10-18 Curevac Ag Vaccine composition
US20190017100A1 (en) 2015-07-01 2019-01-17 Curevac Ag Method for analysis of an rna molecule
WO2017009376A1 (fr) 2015-07-13 2017-01-19 Curevac Ag Procédé de production d'arn à partir d'adn circulaire et adn matriciel correspondant
US20200085852A1 (en) 2015-08-05 2020-03-19 Curevac Ag Epidermal mrna vaccine
WO2017025120A1 (fr) 2015-08-07 2017-02-16 Curevac Ag Procédé pour la production in vivo d'arn dans une cellule hôte
EP3334828B1 (fr) 2015-08-10 2020-10-28 CureVac Real Estate GmbH Procédé d'augmentation de la réplication d'une molécule d'adn circulaire
SG10201913629VA (en) 2015-08-28 2020-03-30 Curevac Ag Artificial nucleic acid molecules
US11225682B2 (en) 2015-10-12 2022-01-18 Curevac Ag Automated method for isolation, selection and/or detection of microorganisms or cells comprised in a solution
MA46317A (fr) * 2015-10-22 2019-08-07 Modernatx Inc Vaccin contre le virus respiratoire syncytial
WO2017081082A2 (fr) 2015-11-09 2017-05-18 Curevac Ag Molécules d'acide nucléique optimisées
US11413346B2 (en) 2015-11-09 2022-08-16 Curevac Ag Rotavirus vaccines
EP3394237A1 (fr) 2015-12-21 2018-10-31 CureVac AG Incrustation pour plaque de culture et procédé de préparation de système de plaque de culture correspondant doté d'une telle incrustation
DK3319622T3 (da) 2015-12-22 2020-05-04 Curevac Ag Fremgangsmåde til fremstilling af rna-molekylesammensætninger
WO2017109161A1 (fr) 2015-12-23 2017-06-29 Curevac Ag Procédé de transcription in vitro d'arn utilisant un tampon contenant un acide dicarboxyliqlue ou un acide tricarboxylique ou un sel de celui-ci
EP3414340B1 (fr) 2016-02-12 2023-08-09 CureVac SE Procédé d'analyse d'arn
US20190049414A1 (en) 2016-02-15 2019-02-14 Curevac Ag Method for analyzing by-products of rna in vitro transcription
BR112018016755A2 (pt) 2016-02-17 2018-12-26 Curevac Ag vacina contra o vírus da zica
US11920174B2 (en) 2016-03-03 2024-03-05 CureVac SE RNA analysis by total hydrolysis and quantification of released nucleosides
WO2017162297A1 (fr) 2016-03-24 2017-09-28 Curevac Ag Pyrophosphatase inorganique immobilisée (ppase)
US20190343942A1 (en) 2016-04-22 2019-11-14 Curevac Ag Rna encoding a tumor antigen
WO2017186928A1 (fr) 2016-04-29 2017-11-02 Curevac Ag Arn codant pour un anticorps
WO2017191264A1 (fr) * 2016-05-04 2017-11-09 Curevac Ag Molécules d'acide nucléique et leurs utilisations
US11078247B2 (en) 2016-05-04 2021-08-03 Curevac Ag RNA encoding a therapeutic protein
WO2017191258A1 (fr) 2016-05-04 2017-11-09 Curevac Ag Vaccins contre l'arnm de la grippe
US20180126003A1 (en) 2016-05-04 2018-05-10 Curevac Ag New targets for rna therapeutics
US20190194760A1 (en) 2016-05-25 2019-06-27 Curevac Ag Novel biomarkers
US11478552B2 (en) 2016-06-09 2022-10-25 Curevac Ag Hybrid carriers for nucleic acid cargo
WO2017212007A1 (fr) 2016-06-09 2017-12-14 Curevac Ag Supports cationiques destinés à l'administration d'acides nucléiques
US20190336611A1 (en) 2016-06-09 2019-11-07 Curevac Ag Hybrid carriers for nucleic acid cargo
EP3424524A3 (fr) 2017-07-04 2019-02-27 CureVac AG Nouvelles molécules d'acide nucléique

Also Published As

Publication number Publication date
US20210401966A1 (en) 2021-12-30
US20190125857A1 (en) 2019-05-02
US11141474B2 (en) 2021-10-12
WO2017191264A1 (fr) 2017-11-09

Similar Documents

Publication Publication Date Title
US20210401966A1 (en) Nucleic acid molecules and uses thereof
US20230226167A1 (en) Henipavirus vaccine
US20230405107A1 (en) Bunyavirales vaccine
US12083190B2 (en) Rabies vaccine
US11786590B2 (en) Rotavirus vaccines
US20240293526A1 (en) Ebolavirus and marburgvirus vaccines
JP7523897B2 (ja) 呼吸器合胞体ウイルス(rsv)ワクチン
US20240024453A1 (en) Zika virus vaccine
US20210162037A1 (en) Influenza mrna vaccines
EP3718565B1 (fr) Vaccins contre le virus respiratoire
US20240131142A1 (en) Flavivirus vaccine
US20190211065A1 (en) High potency immunogenic compositions
EP3558354A1 (fr) Vaccin contre le virus de lassa
EP3035961A1 (fr) Vaccin antirabique

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20181030

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

PUAG Search results despatched under rule 164(2) epc together with communication from examining division

Free format text: ORIGINAL CODE: 0009017

17Q First examination report despatched

Effective date: 20191205

B565 Issuance of search results under rule 164(2) epc

Effective date: 20191205

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 14/005 20060101AFI20191202BHEP

RIN1 Information on inventor provided before grant (corrected)

Inventor name: SCHMIDT, KIM ELLEN

Inventor name: PETSCH, BENJAMIN

Inventor name: RAUCH, SUSANNE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: CUREVAC AG

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

RAP3 Party data changed (applicant data changed or rights of an application transferred)

Owner name: CUREVAC SE

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20230927

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

INTC Intention to grant announced (deleted)
GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20240315

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTC Intention to grant announced (deleted)
INTG Intention to grant announced

Effective date: 20240809